1
|
Parker LE, Papanicolaou KN, Zalesak-Kravec S, Weinberger EM, Kane MA, Foster DB. Retinoic acid signaling and metabolism in heart failure. Am J Physiol Heart Circ Physiol 2025; 328:H792-H813. [PMID: 39933792 DOI: 10.1152/ajpheart.00871.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/24/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Nearly 70 years after studies first showed that the offspring of vitamin A (retinol, ROL)-deficient rats exhibit structural cardiac defects and over 20 years since the role of vitamin A's potent bioactive metabolite hormone, all-trans retinoic acid (ATRA), was elucidated in embryonic cardiac development, the role of the vitamin A metabolites, or retinoids, in adult heart physiology and heart and vascular disease, remains poorly understood. Studies have shown that low serum levels of retinoic acid correlate with higher all-cause and cardiovascular mortality, though the relationship between circulating retinol and ATRA levels, cardiac tissue ATRA levels, and intracellular cardiac ATRA signaling in the context of heart and vascular disease has only begun to be addressed. We have recently shown that patients with idiopathic dilated cardiomyopathy show a nearly 40% decline of in situ cardiac ATRA levels, despite adequate local stores of retinol. Moreover, we and others have shown that the administration of ATRA forestalls the development of heart failure (HF) in rodent models. In this review, we summarize key facets of retinoid metabolism and signaling and discuss mechanisms by which impaired ATRA signaling contributes to several HF hallmarks including hypertrophy, contractile dysfunction, poor calcium handling, redox imbalance, and fibrosis. We highlight unresolved issues in cardiac ATRA metabolism whose pursuit will help refine therapeutic strategies aimed at restoring ATRA homeostasis.
Collapse
Affiliation(s)
- Lauren E Parker
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Kyriakos N Papanicolaou
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | | | - Eva M Weinberger
- School of Medicine, Imperial College London, London, United Kingdom
| | - Maureen A Kane
- School of Pharmacy, University of Maryland, Baltimore, Maryland, United States
| | - D Brian Foster
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
2
|
Basavarajappa D, Chitranshi N, Oddin Mirshahvaladi SS, Gupta VB, Palanivel V, Parrilla GE, Salkar A, Mirzaei M, Komáromy AM, Krezel W, Graham SL, Gupta V. Retinoid X receptor agonist 9CDHRA mitigates retinal ganglion cell apoptosis and neuroinflammation in a mouse model of glaucoma. FASEB J 2025; 39:e70465. [PMID: 40079201 PMCID: PMC11904862 DOI: 10.1096/fj.202402642r] [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: 10/30/2024] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Glaucoma, a leading cause of irreversible blindness, is characterized by the progressive loss of retinal ganglion cells (RGCs) and optic nerve damage, often associated with elevated intraocular pressure (IOP). Retinoid X receptors (RXRs) are ligand-activated transcription factors crucial for neuroprotection, as they regulate gene expression to promote neuronal survival via several biochemical networks and reduce neuroinflammation. This study investigated the therapeutic potential of 9-cis-13,14-dihydroretinoic acid (9CDHRA), an endogenous retinoid RXR agonist, in mitigating RGC degeneration in a high-IOP-induced experimental model of glaucoma. We administered 9CDHRA to glaucomatous mice eyes via intravitreal injections and assessed its effects on endoplasmic reticulum (ER) stress markers, glial cell activation, and RGC survival. Our findings demonstrated that 9CDHRA treatment significantly protected inner retinal function and retinal laminar structure in high-IOP glaucoma. The treatment reduced ER stress markers, increased protein lysine acetylation, and diminished glial cell activation, leading to a significant decrease in apoptotic cells under glaucomatous conditions. These results suggest that 9CDHRA exerts neuroprotective effects by modulating key pathogenic pathways in glaucoma, highlighting its potential as a novel therapeutic strategy for preserving vision in glaucoma.
Collapse
Affiliation(s)
- Devaraj Basavarajappa
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Seyed Shahab Oddin Mirshahvaladi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Veer B Gupta
- School of Medicine, Deakin University, Geelong, Victoria, Australia
| | - Viswanthram Palanivel
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Gabriella E Parrilla
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Akanksha Salkar
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - András M Komáromy
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Wojciech Krezel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut de la Santé et de la Recherche Médicale (U1258), Centre National de la Recherche Scientifique (UMR7104), Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg, Illkirch, France
| | - Stuart L Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Gorodetska I, Lukiyanchuk V, Gawin M, Sliusar M, Linge A, Lohaus F, Hölscher T, Erdmann K, Fuessel S, Borkowetz A, Wojakowska A, Fochtman D, Reardon M, Choudhury A, Antonelli Y, Leal-Egaña A, Köseer AS, Kahya U, Püschel J, Petzold A, Klusa D, Peitzsch C, Kronstein-Wiedemann R, Tonn T, Marczak L, Thomas C, Widłak P, Pietrowska M, Krause M, Dubrovska A. Blood-based detection of MMP11 as a marker of prostate cancer progression regulated by the ALDH1A1-TGF-β1 signaling mechanism. J Exp Clin Cancer Res 2025; 44:105. [PMID: 40122809 PMCID: PMC11931756 DOI: 10.1186/s13046-025-03299-6] [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: 07/25/2024] [Accepted: 01/12/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the second most common type of tumor diagnosed in men and the fifth leading cause of cancer-related death in male patients. The response of metastatic disease to standard treatment is heterogeneous. As for now, there is no curative treatment option available for metastatic PCa, and the clinical tests capable of predicting metastatic dissemination and metastatic response to the therapies are lacking. Our recent study identified aldehyde dehydrogenases ALDH1A1 and ALDH1A3 as critical regulators of PCa metastases. Still, the exact mechanisms mediating the role of these proteins in PCa metastatic dissemination remain not fully understood, and plasma-based biomarkers of these metastatic mechanisms are not available. METHODS Genetic silencing, gene overexpression, or treatment with different concentrations of the retinoic acid (RA) isomers, which are the products of ALDH catalytic activity, were used to modulate the interplay between retinoic acid receptors (RARs) and androgen receptor (AR). RNA sequencing (RNAseq), reporter gene assays, and chromatin immunoprecipitation (ChIP) analysis were employed to validate the role of RARs and AR in the regulation of the transforming growth factor-beta 1 (TGFB1) expression. Gene expression levels of ALDH1A1, ALDH1A3, and the matrix metalloproteinase 11 (MMP11) and their correlation with pathological parameters and clinical outcomes were analysed by mining several publicly available patient datasets as well as our multi-center transcriptomic dataset from patients with high-risk and locally advanced PCa. The level of MMP11 protein was analysed by enzyme-linked immunosorbent assay (ELISA) in independent cohorts of plasma samples from patients with primary or metastatic PCa and healthy donors, while plasma proteome profiles were obtained for selected subsets of PCa patients. RESULTS We could show that ALDH1A1 and ALDH1A3 genes differently regulate TGFB1 expression in a RAR- and AR-dependent manner. We further observed that the TGF-β1 pathway contributes to the regulation of the MMPs, including MMP11. We have confirmed the relevance of MMP11 as a promising clinical marker for PCa using several independent gene expression datasets. Further, we have validated plasma MMP11 level as a prognostic biomarker in patients with metastatic PCa. Finally, we proposed a hypothetical ALDH1A1/MMP11-related plasma proteome-based prognostic signature. CONCLUSIONS TGFB1/MMP11 signaling contributes to the ALDH1A1-driven PCa metastases. MMP11 is a promising blood-based biomarker of PCa progression.
Collapse
Affiliation(s)
- Ielizaveta Gorodetska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Vasyl Lukiyanchuk
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Marta Gawin
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Myroslava Sliusar
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Annett Linge
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Fabian Lohaus
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Tobias Hölscher
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Kati Erdmann
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Susanne Fuessel
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Angelika Borkowetz
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Anna Wojakowska
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Daniel Fochtman
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Mark Reardon
- Division of Cancer Sciences, Translational Radiobiology Group, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, Translational Radiobiology Group, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Yasmin Antonelli
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, Heidelberg, Germany
| | - Aldo Leal-Egaña
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, Heidelberg, Germany
| | - Ayse Sedef Köseer
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Uğur Kahya
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Jakob Püschel
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
| | - Andrea Petzold
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
| | - Daria Klusa
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Claudia Peitzsch
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Romy Kronstein-Wiedemann
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Torsten Tonn
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Lukasz Marczak
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Christian Thomas
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Piotr Widłak
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, Poland
| | - Monika Pietrowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Anna Dubrovska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany.
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.
| |
Collapse
|
4
|
Zhang H, Sáenz de Urturi D, Fernández-Tussy P, Huang Y, Jovin DG, Zhang X, Huang S, Lek M, da Silva Catarino J, Sternak M, Citrin KM, Swirski FK, Gustafsson JÅ, Greif DM, Esplugues E, Biwer LA, Suárez Y, Fernández-Hernando C. Hypercholesterolemia-induced LXR signaling in smooth muscle cells contributes to vascular lesion remodeling and visceral function. Proc Natl Acad Sci U S A 2025; 122:e2417512122. [PMID: 40035761 PMCID: PMC11912459 DOI: 10.1073/pnas.2417512122] [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: 08/29/2024] [Accepted: 01/23/2025] [Indexed: 03/06/2025] Open
Abstract
Vascular smooth muscle cells (VSMC) are the most abundant cell type in the artery's media layer and regulate vascular tone and lesion remodeling during atherogenesis. Like monocyte-derived macrophages, VSMCs accumulate excess lipids and contribute to the total intimal foam cell population in human coronary plaques and mouse aortic atheroma. While there are extensive studies characterizing the contribution of lipid metabolism in macrophage immunometabolic responses in atherosclerotic plaques, the role of VSMC lipid metabolism in regulating vascular function and lesion remodeling in vivo remains poorly understood. Here, we report that the liver X receptor (LXR) signaling pathway in VSMC is continuously activated during atherogenesis. Notably, we found that LXR deficiency in SMCs under hypercholesterolemic conditions influenced lesion remodeling by altering the fate of dedifferentiated SMCs and promoting the accumulation of VSMC-derived transitional cells. This phenotypic switching was accompanied by reduced indices of plaque stability, characterized by a larger necrotic core area and reduced fibrous cap thickness. Moreover, SMC-specific LXR deficiency impaired vascular function and caused visceral myopathy characterized by maladaptive bladder remodeling and gut lipid malabsorption. Mechanistically, we found that the expression of several genes involved in cholesterol efflux and FA synthesis including Abca1, Srebf1, Scd1, Scd2, Acsl3, and Mid1ip1 was downregulated in mice lacking LXRαβ in SMCs, likely contributing to the phenotypic switching of VSMC in the atherosclerotic lesions.
Collapse
MESH Headings
- Liver X Receptors/metabolism
- Liver X Receptors/genetics
- Animals
- Mice
- Hypercholesterolemia/metabolism
- Hypercholesterolemia/pathology
- Hypercholesterolemia/genetics
- Signal Transduction
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Humans
- Vascular Remodeling
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Lipid Metabolism
- Mice, Knockout
- Male
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter 1/genetics
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Hanming Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
| | - Diego Sáenz de Urturi
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
| | - Pablo Fernández-Tussy
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
| | - Yan Huang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
| | - Daniel G. Jovin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT06511
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
| | - Shushu Huang
- Deparment of Genetics, Yale University School of Medicine, New Haven, CT06510
| | - Monkol Lek
- Deparment of Genetics, Yale University School of Medicine, New Haven, CT06510
| | | | - Magdalena Sternak
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Bobrzynskiego 14, 30-348, Krakow, Poland
| | - Kathryn M. Citrin
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
- Department of Cellular & Molecular Physiology, Yale University, New Haven, CT06510
| | - Fillip K. Swirski
- Caridovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center of Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX77204
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT06511
- Deparment of Genetics, Yale University School of Medicine, New Haven, CT06510
| | - Enric Esplugues
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
| | - Lauren A. Biwer
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
- Department of Pathology, Yale University School of Medicine, New Haven, CT06520
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT06520
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT06520
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT06520
- Department of Pathology, Yale University School of Medicine, New Haven, CT06520
| |
Collapse
|
5
|
Kakiyama G, Bai-Kamara N, Rodriguez-Agudo D, Takei H, Minowa K, Fuchs M, Biddinger S, Windle JJ, Subler MA, Murai T, Suzuki M, Nittono H, Sanyal A, Pandak WM. Liver specific transgenic expression of CYP7B1 attenuates early western diet-induced MASLD progression. J Lipid Res 2025; 66:100757. [PMID: 39952566 PMCID: PMC11954105 DOI: 10.1016/j.jlr.2025.100757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
Effect of liver specific oxysterol 7α-hydroxylase (CYP7B1) overexpression on the Western diet (WD)-induced metabolic dysfunction-associated steatotic liver disease (MASLD) progression was studied in mice. Among various hepatic genes impacted during MASLD development, CYP7B1 is consistently suppressed in multiple MASLD mouse models and in human MASLD cohorts. CYP7B1 enzyme suppression leads to accumulations of bioactive oxysterols such as (25R)26-hydroxycholesterol (26HC) and 25-hydroxycholesterol (25HC). We challenged liver specific CYP7B1 transgenic (CYP7B1hep.tg) overexpressing mice with ad libitum WD feeding. Unlike their WT counterparts, WD-fed CYP7B1hep.tg mice developed no significant hepatotoxicity as evidenced by liver histology, lipid quantifications, and serum biomarker analyses. Hepatic 26HC and 25HC levels were maintained at the basal levels. The comparative gene expression/lipidomic analyses between WT and CYP7B1hep.tg mice revealed that chronically accumulated 26HC initiates LXR/PPAR-mediated hepatic fatty acid uptake and lipogenesis which surpasses fatty acid metabolism and export; compromising metabolic functions. In addition, major pathways related to oxidative stress, inflammation, and immune system including retinol metabolism, arachidonic acid metabolism, and linoleic acid metabolism were significantly impacted in the WD-fed WT mice. All pathways were unaltered in CYP7B1hep.tg mice liver. Furthermore, the nucleus of WT mouse liver but not of CYP7B1hep.tg mouse liver accumulated 26HC and 25HC in response to WD. These data strongly suggested that these two oxysterols are specifically important in nuclear transcriptional regulation for the described cytotoxic pathways. In conclusion, this study represents a "proof-of-concept" that maintaining normal mitochondrial cholesterol metabolism with hepatic CYP7B1 expression prevents oxysterol-driven liver toxicity; thus attenuating MASLD progression.
Collapse
Affiliation(s)
- Genta Kakiyama
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Health Care System, Richmond, VA, USA.
| | - Nanah Bai-Kamara
- Research Services, Central Virginia Veterans Affairs Health Care System, Richmond, VA, USA
| | - Daniel Rodriguez-Agudo
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Health Care System, Richmond, VA, USA
| | - Hajime Takei
- Junshin Clinic Bile Acid Institute, Meguro-ku, Tokyo, Japan
| | - Kei Minowa
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Health Care System, Richmond, VA, USA; Department of Pediatrics, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Michael Fuchs
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Health Care System, Richmond, VA, USA
| | - Sudha Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, MA, USA
| | - Jolene J Windle
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond VA, USA
| | - Mark A Subler
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond VA, USA
| | - Tsuyoshi Murai
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari, Hokkaido, Japan
| | - Mitsuyoshi Suzuki
- Department of Pediatrics, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | | | - Arun Sanyal
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - William M Pandak
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA; Research Services, Central Virginia Veterans Affairs Health Care System, Richmond, VA, USA
| |
Collapse
|
6
|
De Felice D, Alaimo A, Bressan D, Genovesi S, Marmocchi E, Annesi N, Beccaceci G, Dalfovo D, Cutrupi F, Medaglia S, Foletto V, Lorenzoni M, Gandolfi F, Kannan S, Verma CS, Vasciaveo A, Shen MM, Romanel A, Chiacchiera F, Cambuli F, Lunardi A. Rarγ-Foxa1 signaling promotes luminal identity in prostate progenitors and is disrupted in prostate cancer. EMBO Rep 2025; 26:443-469. [PMID: 39633177 PMCID: PMC11772605 DOI: 10.1038/s44319-024-00335-y] [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: 04/16/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Retinoic acid (RA) signaling is a master regulator of vertebrate development with crucial roles in body axis orientation and tissue differentiation, including in the reproductive system. However, a mechanistic understanding of how RA signaling governs cell lineage identity is often missing. Here, leveraging prostate organoid technology, we show that RA signaling orchestrates the commitment of adult mouse prostate progenitors to glandular identity, epithelial barrier integrity, and specification of prostatic lumen. RA-dependent RARγ activation promotes the expression of Foxa1, which synergizes with the androgen pathway for luminal expansion, cytoarchitecture and function. FOXA1 mutations are common in prostate and breast cancers, though their pathogenic mechanism is incompletely understood. Combining functional genetics with structural modeling of FOXA1 folding and chromatin binding analyses, we discover that FOXA1F254E255 is a loss-of-function mutation compromising its transcriptional function and luminal fate commitment of prostate progenitors. Overall, we define RA as an instructive signal for glandular identity in adult prostate progenitors. Importantly, we identify cancer-associated FOXA1 indels affecting residue F254 as loss-of-function mutations promoting dedifferentiation of adult prostate progenitors.
Collapse
Affiliation(s)
- Dario De Felice
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Alessandro Alaimo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Davide Bressan
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Sacha Genovesi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Elisa Marmocchi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Nicole Annesi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Giulia Beccaceci
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Davide Dalfovo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Federico Cutrupi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Stefano Medaglia
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Veronica Foletto
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Marco Lorenzoni
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Francesco Gandolfi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Srinivasaraghavan Kannan
- Bioinformatics Institute (Agency for Science, Technology and Research, A*STAR), 30 Biopolis Street, 07-01 Matrix, Singapore, 138671, Singapore
| | - Chandra S Verma
- Bioinformatics Institute (Agency for Science, Technology and Research, A*STAR), 30 Biopolis Street, 07-01 Matrix, Singapore, 138671, Singapore
- Department of Biological Sciences, National University of Singapore, 14 Science Drive, Singapore, 117543, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Alessandro Vasciaveo
- Departments of Medicine, Genetics & Development, Urology and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Michael M Shen
- Departments of Medicine, Genetics & Development, Urology and Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Alessandro Romanel
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Fulvio Chiacchiera
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy
| | - Francesco Cambuli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy.
- Human Technopole, via Rita Levi Montalcini 1, Milan, Italy.
| | - Andrea Lunardi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, TN, Italy.
| |
Collapse
|
7
|
Feng Q, Su C, Yang C, Wu M, Li X, Lin X, Zeng Y, He J, Wang Y, Guo L, Wen C, Cai F, Zhang J, Fan X, Guan M. RXRα/MR signaling promotes diabetic kidney disease by facilitating renal tubular epithelial cells senescence and metabolic reprogramming. Transl Res 2024; 274:101-117. [PMID: 39424127 DOI: 10.1016/j.trsl.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
Cell senescence and metabolic reprogramming are significant features of diabetic kidney disease (DKD). However, the underlying mechanisms between cell senescence and metabolic reprogramming are poorly defined. Here, we report that retinoid X receptor α (RXRα), a key nuclear receptor transcription factor, regulates cell senescence and metabolic reprogramming in DKD. Through high-throughput sequencing, bioinformatic analysis and experimental validation, we confirmed the critical role of RXRα in promoting cell senescence and metabolic dysregulation in renal tubular epithelial cells (RTECs) induced by lipid overload. In vivo, in situ injection of AAV9-shRxra into the kidney reduced proteinuria, RTECs senescence and insulin resistance in DKD mice. In vitro, knockdown of RXRα markedly improved G2/M phase arrest and suppressed the expression of senescence-associated secretory phenotypes (SASPs). Protein-protein interaction (PPI) analysis and unbiased bioinformatics were employed to identify the direct interactions between RXRα and the mineralocorticoid receptor (MR), which were subsequently validated through coimmunoprecipitation. Gene network analysis revealed the collaborative regulatory role of RXRα and MR in RTECs senescence. In an accelerated aging mouse model, treatment with a MR antagonist has been shown to inhibite the RXRα/MR signaling, improve RTECs senescence, and reduce interstitial fibrosis and lipid deposition in the kidneys. These findings indicate that inhibition of RXRα/MR signaling could alleviate cell senescence during metabolic disorders. Thus, our study revealed that RXRα/MR signaling serves as a critical regulatory factor mediating the crosstalk between cell senescence and metabolic reprogramming, shedding light on a novel mechanism for targeting cell senescence and metabolic dysregulation in DKD.
Collapse
Affiliation(s)
- Qijian Feng
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515; Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, PR China
| | - Chang Su
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Chuyi Yang
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Minghai Wu
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Xuelin Li
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Xiaochun Lin
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Yanmei Zeng
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Jintao He
- School of Medicine, South China University of Technology, Guangzhou 510080, PR China
| | - Yuan Wang
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Lei Guo
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Churan Wen
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Feifei Cai
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Jin Zhang
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Xinzhao Fan
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515
| | - Meiping Guan
- Department of Endocrinology & Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China. 510515.
| |
Collapse
|
8
|
Balbuena E, Milhem F, Kiremitci BZ, Williams TI, Collins L, Shu Q, Eroglu A. The biochemical effects of carotenoids in orange carrots on the colonic proteome in a mouse model of diet-induced obesity. Front Nutr 2024; 11:1492380. [PMID: 39588046 PMCID: PMC11587903 DOI: 10.3389/fnut.2024.1492380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024] Open
Abstract
Introduction Carotenoids are naturally occurring pigments in plants and are responsible for the orange, yellow, and red color of fruits and vegetables. Carrots are one of the primary dietary sources of carotenoids. The biological activities of carotenoids in higher organisms, including their immunomodulatory activities, are well documented in most tissues but not the large intestine. The gastrointestinal barrier acts as a line of defense against the systemic invasion of pathogenic bacteria, especially at the colonic level. Methods To test whether carotenoids in orange carrots can alleviate obesity-associated gut inflammation and strengthen the intestinal barrier function, male C57BL/6J mice were randomized to one of four experimental diets for 20 weeks (n = 20 animals/group): Low-fat diet (LFD, 10% calories from fat), high-fat diet (HFD, 45% calories from fat), HFD with white carrot powder (HFD+WC), or HFD with orange carrot powder (HFD + OC). Colon tissues were harvested to analyze the biochemical effects of carotenoids in carrots. The distal sections were subjected to isobaric labeling-based quantitative proteomics in which tryptic peptides were labeled with tandem mass tags, followed by fractionation and LC-MS/MS analysis in an Orbitrap Eclipse Tribrid instrument. Results High-performance liquid chromatography results revealed that the HFD+WC pellets were carotenoid-deficient, and the HFD+OC pellets contained high concentrations of provitamin A carotenoids, specifically α-carotene and β-carotene. As a result of the quantitative proteomics, a total of 4410 differentially expressed proteins were identified. Intestinal barrier-associated proteins were highly upregulated in the HFD+OC group, particularly mucin-2 (MUC-2). Upon closer investigation into mucosal activity, other proteins related to MUC-2 functionality and tight junction management were upregulated by the HFD+OC dietary intervention. Discussion Collectively, our findings suggest that carotenoid-rich foods can prevent high-fat diet-induced intestinal barrier disruption by promoting colonic mucus synthesis and secretion in mammalian organisms. Data are available via ProteomeXchange with identifier PXD054150.
Collapse
Affiliation(s)
- Emilio Balbuena
- Department of Molecular and Structural Biochemistry, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, United States
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| | - Fadia Milhem
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| | - Buse Zeren Kiremitci
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| | - Taufika Islam Williams
- Department of Chemistry, North Carolina State University, Raleigh, NC, United States
- Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, NC, United States
| | - Leonard Collins
- Department of Chemistry, North Carolina State University, Raleigh, NC, United States
- Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, NC, United States
| | - Qingbo Shu
- Molecular Education, Technology and Research Innovation Center (METRIC), North Carolina State University, Raleigh, NC, United States
| | - Abdulkerim Eroglu
- Department of Molecular and Structural Biochemistry, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, United States
- Plants for Human Health Institute, North Carolina State University, Kannapolis, NC, United States
| |
Collapse
|
9
|
Engfer ZJ, Palczewski K. The multifaceted roles of retinoids in eye development, vision, and retinal degenerative diseases. Curr Top Dev Biol 2024; 161:235-296. [PMID: 39870435 DOI: 10.1016/bs.ctdb.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Vitamin A (all-trans-retinol; at-Rol) and its derivatives, known as retinoids, have been adopted by vertebrates to serve as visual chromophores and signaling molecules, particularly in the eye/retina. Few tissues rely on retinoids as heavily as the retina, and the study of genetically modified mouse models with deficiencies in specific retinoid-metabolizing proteins has allowed us to gain insight into the unique or redundant roles of these proteins in at-Rol uptake and storage, or their downstream roles in retinal development and function. These processes occur during embryogenesis and continue throughout life. This review delves into the role of these genes in supporting retinal function and maps the impact that genetically modified mouse models have had in studying retinoid-related genes. These models display distinct perturbations in retinoid biochemistry, physiology, and metabolic flux, mirroring human ocular diseases.
Collapse
Affiliation(s)
- Zachary J Engfer
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States.
| | - Krzysztof Palczewski
- Center for Translational Vision Research, Department of Ophthalmology, Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, United States; Department of Chemistry, University of California Irvine, Irvine, CA, United States; Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
10
|
Xu R, Zhang L, Pan H, Zhang Y. Retinoid X receptor heterodimers in hepatic function: structural insights and therapeutic potential. Front Pharmacol 2024; 15:1464655. [PMID: 39478961 PMCID: PMC11521896 DOI: 10.3389/fphar.2024.1464655] [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: 07/14/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
Nuclear receptors (NRs) are key regulators of multiple physiological functions and pathological changes in the liver in response to a variety of extracellular signaling changes. Retinoid X receptor (RXR) is a special member of the NRs, which not only responds to cellular signaling independently, but also regulates multiple signaling pathways by forming heterodimers with various other NR. Therefore, RXR is widely involved in hepatic glucose metabolism, lipid metabolism, cholesterol metabolism and bile acid homeostasis as well as hepatic fibrosis. Specific activation of particular dimers regulating physiological and pathological processes may serve as important pharmacological targets. So here we describe the basic information and structural features of the RXR protein and its heterodimers, focusing on the role of RXR heterodimers in a number of physiological processes and pathological imbalances in the liver, to provide a theoretical basis for RXR as a promising drug target.
Collapse
Affiliation(s)
- Renjie Xu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linyue Zhang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Pan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Tong X, Remsik J, Brook J, Petrova B, Xu L, Li MJ, Snyder J, Chabot K, Estrera R, Osei-Gyening I, Nobre AR, Wang H, Osman AM, Wong AYL, Sidharta M, Piedrafita-Ortiz S, Manoranjan B, Zhou T, Murali R, Hamard PJ, Koche R, He Y, Kanarek N, Boire A. Retinoid X Receptor Signaling Mediates Cancer Cell Lipid Metabolism in the Leptomeninges. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618083. [PMID: 39464048 PMCID: PMC11507812 DOI: 10.1101/2024.10.13.618083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Cancer cells metastatic to the leptomeninges encounter a metabolically-challenging extreme microenvironment. To understand adaptations to this space, we subjected leptomeningeal-metastatic (LeptoM) mouse breast and lung cancers isolated from either the leptomeninges or orthotopic primary sites to ATAC-and RNA-sequencing. When inhabiting the leptomeninges, the LeptoM cells demonstrated transcription downstream of retinoid-X-receptors (RXRs). We found evidence of local retinoic acid (RA) generation in both human leptomeningeal metastasis and mouse models in the form of elevated spinal fluid retinol and expression of RA-generating dehydrogenases within the leptomeningeal microenvironment. Stimulating LeptoM cells with RA induced expression of transcripts encoding de novo fatty acid synthesis pathway enzymes in vitro . In vivo , while deletion of Stra6 did not alter cancer cell leptomeningeal growth, knockout of Rxra/b/g interrupted cancer cell lipid biosynthesis and arrested cancer growth. These observations illustrate a mechanism whereby metastatic cancer cells awake locally-generated developmental cues for metabolically reprograming, suggesting novel therapeutic approaches.
Collapse
|
12
|
Esposito M, Amory JK, Kang Y. The pathogenic role of retinoid nuclear receptor signaling in cancer and metabolic syndromes. J Exp Med 2024; 221:e20240519. [PMID: 39133222 PMCID: PMC11318670 DOI: 10.1084/jem.20240519] [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: 05/23/2024] [Revised: 07/13/2024] [Accepted: 07/26/2024] [Indexed: 08/13/2024] Open
Abstract
The retinoid nuclear receptor pathway, activated by the vitamin A metabolite retinoic acid, has been extensively investigated for over a century. This study has resulted in conflicting hypotheses about how the pathway regulates health and how it should be pharmaceutically manipulated. These disagreements arise from a fundamental contradiction: retinoid agonists offer clear benefits to select patients with rare bone growth disorders, acute promyelocytic leukemia, and some dermatologic diseases, yet therapeutic retinoid pathway activation frequently causes more harm than good, both through acute metabolic dysregulation and a delayed cancer-promoting effect. In this review, we discuss controlled clinical, mechanistic, and genetic data to suggest several disease settings where inhibition of the retinoid pathway may be a compelling therapeutic strategy, such as solid cancers or metabolic syndromes, and also caution against continued testing of retinoid agonists in cancer patients. Considerable evidence suggests a central role for retinoid regulation of immunity and metabolism, with therapeutic opportunities to antagonize retinoid signaling proposed in cancer, diabetes, and obesity.
Collapse
Affiliation(s)
- Mark Esposito
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Kayothera, Inc , Seattle, WA, USA
| | | | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Institute for Cancer Research Princeton Branch , Princeton, NJ, USA
| |
Collapse
|
13
|
Stewart KS, Abdusselamoglu MD, Tierney MT, Gola A, Hur YH, Gonzales KAU, Yuan S, Bonny AR, Yang Y, Infarinato NR, Cowley CJ, Levorse JM, Pasolli HA, Ghosh S, Rothlin CV, Fuchs E. Stem cells tightly regulate dead cell clearance to maintain tissue fitness. Nature 2024; 633:407-416. [PMID: 39169186 PMCID: PMC11390485 DOI: 10.1038/s41586-024-07855-6] [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: 04/30/2023] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Billions of cells are eliminated daily from our bodies1-4. Although macrophages and dendritic cells are dedicated to migrating and engulfing dying cells and debris, many epithelial and mesenchymal tissue cells can digest nearby apoptotic corpses1-4. How these non-motile, non-professional phagocytes sense and eliminate dying cells while maintaining their normal tissue functions is unclear. Here we explore the mechanisms that underlie their multifunctionality by exploiting the cyclical bouts of tissue regeneration and degeneration during hair cycling. We show that hair follicle stem cells transiently unleash phagocytosis at the correct time and place through local molecular triggers that depend on both lipids released by neighbouring apoptotic corpses and retinoids released by healthy counterparts. We trace the heart of this dual ligand requirement to RARγ-RXRα, whose activation enables tight regulation of apoptotic cell clearance genes and provides an effective, tunable mechanism to offset phagocytic duties against the primary stem cell function of preserving tissue integrity during homeostasis. Finally, we provide functional evidence that hair follicle stem cell-mediated phagocytosis is not simply redundant with professional phagocytes but rather has clear benefits to tissue fitness. Our findings have broad implications for other non-motile tissue stem or progenitor cells that encounter cell death in an immune-privileged niche.
Collapse
Affiliation(s)
- Katherine S Stewart
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA.
| | - Merve Deniz Abdusselamoglu
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Matthew T Tierney
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Anita Gola
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Yun Ha Hur
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Kevin A U Gonzales
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Department of Discovery Technology and Genomics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Shaopeng Yuan
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge, UK
| | - Alain R Bonny
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Yihao Yang
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Altos Labs, San Diego, CA, USA
| | - Nicole R Infarinato
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- PrecisionScienta, Yardley, PA, USA
| | - Christopher J Cowley
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John M Levorse
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
- Cardiovascular Research Group, Temple University, Philadelphia, PA, USA
| | - Hilda Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Sourav Ghosh
- Departments of Neurology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Carla V Rothlin
- Departments of Immunobiology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
14
|
Chen Y, Anderson MT, Payne N, Santori FR, Ivanova NB. Nuclear Receptors and the Hidden Language of the Metabolome. Cells 2024; 13:1284. [PMID: 39120315 PMCID: PMC11311682 DOI: 10.3390/cells13151284] [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: 06/12/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Nuclear hormone receptors (NHRs) are a family of ligand-regulated transcription factors that control key aspects of development and physiology. The regulation of NHRs by ligands derived from metabolism or diet makes them excellent pharmacological targets, and the mechanistic understanding of how NHRs interact with their ligands to regulate downstream gene networks, along with the identification of ligands for orphan NHRs, could enable innovative approaches for cellular engineering, disease modeling and regenerative medicine. We review recent discoveries in the identification of physiologic ligands for NHRs. We propose new models of ligand-receptor co-evolution, the emergence of hormonal function and models of regulation of NHR specificity and activity via one-ligand and two-ligand models as well as feedback loops. Lastly, we discuss limitations on the processes for the identification of physiologic NHR ligands and emerging new methodologies that could be used to identify the natural ligands for the remaining 17 orphan NHRs in the human genome.
Collapse
Affiliation(s)
- Yujie Chen
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; (Y.C.); (M.T.A.); (N.P.)
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Matthew Tom Anderson
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; (Y.C.); (M.T.A.); (N.P.)
| | - Nathaniel Payne
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; (Y.C.); (M.T.A.); (N.P.)
| | - Fabio R. Santori
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; (Y.C.); (M.T.A.); (N.P.)
| | - Natalia B. Ivanova
- Center for Molecular Medicine, University of Georgia, Athens, GA 30602, USA; (Y.C.); (M.T.A.); (N.P.)
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
15
|
Steinhoff JS, Wagner C, Dähnhardt HE, Košić K, Meng Y, Taschler U, Pajed L, Yang N, Wulff S, Kiefer MF, Petricek KM, Flores RE, Li C, Dittrich S, Sommerfeld M, Guillou H, Henze A, Raila J, Wowro SJ, Schoiswohl G, Lass A, Schupp M. Adipocyte HSL is required for maintaining circulating vitamin A and RBP4 levels during fasting. EMBO Rep 2024; 25:2878-2895. [PMID: 38769419 PMCID: PMC11239848 DOI: 10.1038/s44319-024-00158-x] [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: 03/27/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024] Open
Abstract
Vitamin A (retinol) is distributed via the blood bound to its specific carrier protein, retinol-binding protein 4 (RBP4). Retinol-loaded RBP4 is secreted into the circulation exclusively from hepatocytes, thereby mobilizing hepatic retinoid stores that represent the major vitamin A reserves in the body. The relevance of extrahepatic retinoid stores for circulating retinol and RBP4 levels that are usually kept within narrow physiological limits is unknown. Here, we show that fasting affects retinoid mobilization in a tissue-specific manner, and that hormone-sensitive lipase (HSL) in adipose tissue is required to maintain serum concentrations of retinol and RBP4 during fasting in mice. We found that extracellular retinol-free apo-RBP4 induces retinol release by adipocytes in an HSL-dependent manner. Consistently, global or adipocyte-specific HSL deficiency leads to an accumulation of retinoids in adipose tissue and a drop of serum retinol and RBP4 during fasting, which affects retinoid-responsive gene expression in eye and kidney and lowers renal retinoid content. These findings establish a novel crosstalk between liver and adipose tissue retinoid stores for the maintenance of systemic vitamin A homeostasis during fasting.
Collapse
Affiliation(s)
- Julia S Steinhoff
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Carina Wagner
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Henriette E Dähnhardt
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Kristina Košić
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Yueming Meng
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Ulrike Taschler
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Laura Pajed
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Na Yang
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Sascha Wulff
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Marie F Kiefer
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Konstantin M Petricek
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Roberto E Flores
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Chen Li
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Sarah Dittrich
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Manuela Sommerfeld
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), INRAE, ENVT, INP- PURPAN, UMR 1331, UPS, Université de Toulouse, Toulouse, France
| | - Andrea Henze
- Martin Luther University Halle-Wittenberg, Institute of Agricultural and Nutritional Sciences, Halle, Germany
- Junior Research Group ProAID, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Jens Raila
- Department of Physiology and Pathophysiology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Sylvia J Wowro
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany
| | - Gabriele Schoiswohl
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Michael Schupp
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Max Rubner Center (MRC) for Cardiovascular-Metabolic-Renal Research, Berlin, Germany.
| |
Collapse
|
16
|
Gautheron J, Elsayed S, Pistorio V, Lockhart S, Zammouri J, Auclair M, Koulman A, Meadows SR, Lhomme M, Ponnaiah M, Si-Bouazza R, Fabrega S, Belkadi A, Delaunay JL, Aït-Slimane T, Fève B, Vigouroux C, Abdel Ghaffar TY, O’Rahilly S, Jéru I. ADH1B, the adipocyte-enriched alcohol dehydrogenase, plays an essential, cell-autonomous role in human adipogenesis. Proc Natl Acad Sci U S A 2024; 121:e2319301121. [PMID: 38838011 PMCID: PMC11181076 DOI: 10.1073/pnas.2319301121] [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: 11/03/2023] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Alcohol dehydrogenase 1B (ADH1B) is a primate-specific enzyme which, uniquely among the ADH class 1 family, is highly expressed both in adipose tissue and liver. Its expression in adipose tissue is reduced in obesity and increased by insulin stimulation. Interference with ADH1B expression has also been reported to impair adipocyte function. To better understand the role of ADH1B in adipocytes, we used CRISPR/Cas9 to delete ADH1B in human adipose stem cells (ASC). Cells lacking ADH1B failed to differentiate into mature adipocytes manifested by minimal triglyceride accumulation and a marked reduction in expression of established adipocyte markers. As ADH1B is capable of converting retinol to retinoic acid (RA), we conducted rescue experiments. Incubation of ADH1B-deficient preadipocytes with 9-cis-RA, but not with all-transretinol, significantly rescued their ability to accumulate lipids and express markers of adipocyte differentiation. A homozygous missense variant in ADH1B (p.Arg313Cys) was found in a patient with congenital lipodystrophy of unknown cause. This variant significantly impaired the protein's dimerization, enzymatic activity, and its ability to rescue differentiation in ADH1B-deficient ASC. The allele frequency of this variant in the Middle Eastern population suggests that it is unlikely to be a fully penetrant cause of severe lipodystrophy. In conclusion, ADH1B appears to play an unexpected, crucial and cell-autonomous role in human adipocyte differentiation by serving as a necessary source of endogenous retinoic acid.
Collapse
Affiliation(s)
- Jérémie Gautheron
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Solaf Elsayed
- Medical Genetics Department, Faculty of Medicine, Ain Shams University, Cairo11566, Egypt
| | - Valeria Pistorio
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Sam Lockhart
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Jamila Zammouri
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Martine Auclair
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Albert Koulman
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Sarah R. Meadows
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Marie Lhomme
- Omics Lipidomics, Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Maharajah Ponnaiah
- Data sciences unit, Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Redouane Si-Bouazza
- Viral Vector and Gene Transfer Platform, Structure Federative de Recherche Necker, Université Paris Cité, Paris75015, France
| | - Sylvie Fabrega
- Viral Vector and Gene Transfer Platform, Structure Federative de Recherche Necker, Université Paris Cité, Paris75015, France
| | - Abdelaziz Belkadi
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha24144, Qatar
| | - Qatar Genome Project
- Qatar Genome Program, Foundation Research, Development and Innovation, Qatar Foundation, Doha24144, Qatar
| | - Jean-Louis Delaunay
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Tounsia Aït-Slimane
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
| | - Bruno Fève
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
- Centre National de Référence des Pathologies Rares de l’Insulino-Sécrétion et de l’Insulino-Sensibilité, Service de Diabétologie et Endocrinologie de la Reproduction, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Paris75012, France
| | - Corinne Vigouroux
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
- Centre National de Référence des Pathologies Rares de l’Insulino-Sécrétion et de l’Insulino-Sensibilité, Service de Diabétologie et Endocrinologie de la Reproduction, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Paris75012, France
| | | | - Stephen O’Rahilly
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Isabelle Jéru
- Centre de Recherche Saint-Antoine, Sorbonne Université-Inserm, Paris75012, France
- Foundation for Innovation in Cardiometabolism and Nutrition, Paris75013, France
- Medical Genetics Unit, Biology, Genomics and Hygiene Medical-University Department, Pitié-Salpêtrière Hospital, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Paris75013, France
| |
Collapse
|
17
|
Rühl R, Bánáti D. Analysis of the current vitamin A terminology and dietary regulations from vitamin A 1 to vitamin A 5. INT J VITAM NUTR RES 2024; 94:326-333. [PMID: 38506673 DOI: 10.1024/0300-9831/a000807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Dietary recommendations on vitamin intake for human food fortification concerning vitamin A in various countries, larger economic zones and international organizations are mainly based on the Food and Agriculture Organization of the United Nations (FAO)/World Health Organization (WHO) "Codex Alimentarius standards". The general vitamin A terminology is based on regulations of the International Union of Pure and Applied Chemistry (IUPAC) that are used to describe the involved derivatives. These regulations and terminology were set up in the middle of the last century. Starting with the decade of the 80ies in the 20th century a large improvement of molecular biological methodologies, background physiological mechanisms as well as analytical techniques contributed to a large diversification of this simply claimed vitamin A terminology. Unfortunately, the following terminology and governmental regulations for food fortification are imprecise and non-harmonized. In this article we tried to unravel this terminology for updating terminology, nutritional suggestions and governmental regulations for vitamin A, which are currently based on various uncertainties. According to the current regulations, the newly found vitamin A5/X can be included in the current vitamin A terminology as "vitamin A5" or alternatively or even in parallel as a new vitamin A-independent terminology as "vitamin X". Based on the detailed knowledge of research from the early beginning of general vitamin A pathway identification towards detailed research of the last decades the commonly used and simplified term vitamin A with relevance for governmental recommendations on vitamin intake and food fortification advice was now more correctly sub-categorized to further vitamin A1, and A5 sub-categories with vitamin A1-alcohol as retinol, vitamin A2-alcohol as 3,4-didehydroretinol and vitamin A5-alcohol as 9-cis-13,14-dihydroretinol as their mainly relevant vitamin forms present in the human organism. Here we suggest and advise how the vitamin A terminology and further governmental regulations should be organized depending on a successful unraveling of the organization of the current vitamin A terminology.
Collapse
Affiliation(s)
| | - Diána Bánáti
- Department of Food Engineering, Faculty of Engineering, University of Szeged, Hungary
| |
Collapse
|
18
|
Loeffler DA. Enhancing of cerebral Abeta clearance by modulation of ABC transporter expression: a review of experimental approaches. Front Aging Neurosci 2024; 16:1368200. [PMID: 38872626 PMCID: PMC11170721 DOI: 10.3389/fnagi.2024.1368200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/01/2024] [Indexed: 06/15/2024] Open
Abstract
Clearance of amyloid-beta (Aβ) from the brain is impaired in both early-onset and late-onset Alzheimer's disease (AD). Mechanisms for clearing cerebral Aβ include proteolytic degradation, antibody-mediated clearance, blood brain barrier and blood cerebrospinal fluid barrier efflux, glymphatic drainage, and perivascular drainage. ATP-binding cassette (ABC) transporters are membrane efflux pumps driven by ATP hydrolysis. Their functions include maintenance of brain homeostasis by removing toxic peptides and compounds, and transport of bioactive molecules including cholesterol. Some ABC transporters contribute to lowering of cerebral Aβ. Mechanisms suggested for ABC transporter-mediated lowering of brain Aβ, in addition to exporting of Aβ across the blood brain and blood cerebrospinal fluid barriers, include apolipoprotein E lipidation, microglial activation, decreased amyloidogenic processing of amyloid precursor protein, and restricting the entrance of Aβ into the brain. The ABC transporter superfamily in humans includes 49 proteins, eight of which have been suggested to reduce cerebral Aβ levels. This review discusses experimental approaches for increasing the expression of these ABC transporters, clinical applications of these approaches, changes in the expression and/or activity of these transporters in AD and transgenic mouse models of AD, and findings in the few clinical trials which have examined the effects of these approaches in patients with AD or mild cognitive impairment. The possibility that therapeutic upregulation of ABC transporters which promote clearance of cerebral Aβ may slow the clinical progression of AD merits further consideration.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, United States
| |
Collapse
|
19
|
Lerner UH. Vitamin A - discovery, metabolism, receptor signaling and effects on bone mass and fracture susceptibility. Front Endocrinol (Lausanne) 2024; 15:1298851. [PMID: 38711977 PMCID: PMC11070503 DOI: 10.3389/fendo.2024.1298851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 04/02/2024] [Indexed: 05/08/2024] Open
Abstract
The first evidence of the existence of vitamin A was the observation 1881 that a substance present in small amounts in milk was necessary for normal development and life. It was not until more than 100 years later that it was understood that vitamin A acts as a hormone through nuclear receptors. Unlike classical hormones, vitamin A cannot be synthesized by the body but needs to be supplied by the food as retinyl esters in animal products and ß-carotene in vegetables and fruits. Globally, vitamin A deficiency is a huge health problem, but in the industrialized world excess of vitamin A has been suggested to be a risk factor for secondary osteoporosis and enhanced susceptibility to fractures. Preclinical studies unequivocally have shown that increased amounts of vitamin A cause decreased cortical bone mass and weaker bones due to enhanced periosteal bone resorption. Initial clinical studies demonstrated a negative association between intake of vitamin A, as well as serum levels of vitamin A, and bone mass and fracture susceptibility. In some studies, these observations have been confirmed, but in other studies no such associations have been observed. One meta-analysis found that both low and high serum levels of vitamin A were associated with increased relative risk of hip fractures. Another meta-analysis also found that low levels of serum vitamin A increased the risk for hip fracture but could not find any association with high serum levels of vitamin A and hip fracture. It is apparent that more clinical studies, including large numbers of incident fractures, are needed to determine which levels of vitamin A that are harmful or beneficial for bone mass and fracture. It is the aim of the present review to describe how vitamin A was discovered and how vitamin A is absorbed, metabolized and is acting as a ligand for nuclear receptors. The effects by vitamin A in preclinical studies are summarized and the clinical investigations studying the effect by vitamin A on bone mass and fracture susceptibility are discussed in detail.
Collapse
Affiliation(s)
- Ulf H. Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
20
|
Felice DD, Alaimo A, Bressan D, Genovesi S, Marmocchi E, Annesi N, Beccaceci G, Dalfovo D, Cutrupi F, Foletto V, Lorenzoni M, Gandolfi F, Kannan S, Verma CS, Vasciaveo A, Shen MM, Romanel A, Chiacchiera F, Cambuli F, Lunardi A. Rarγ -Foxa1 signaling promotes luminal identity in prostate progenitors and is disrupted in prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.06.583256. [PMID: 38496627 PMCID: PMC10942448 DOI: 10.1101/2024.03.06.583256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Retinoic acid (RA) signaling is a master regulator of vertebrate development with crucial roles in directing body axis orientation and tissue differentiation, including in the reproductive system. However, a mechanistic understanding of how RA signaling promotes cell lineage identity in different tissues is often missing. Here, leveraging prostate organoid technology, we demonstrated that RA signaling orchestrates the commitment of adult mouse prostate progenitors to glandular identity, epithelial barrier integrity, and ultimately, proper specification of the prostatic lumen. Mechanistically, RA-dependent RARγ activation promotes the expression of the pioneer factor Foxa1, which synergizes with the androgen pathway for proper luminal expansion, cytoarchitecture and function. FOXA1 nucleotide variants are common in human prostate and breast cancers and considered driver mutations, though their pathogenic mechanism is incompletely understood. Combining functional genetics experiments with structural modeling of FOXA1 folding and chromatin binding analyses, we discovered that FOXA1 F254E255 is a loss-of-function mutation leading to compromised transcriptional function and lack of luminal fate commitment of prostate progenitors. Overall, we define RA as a crucial instructive signal for glandular identity in adult prostate progenitors. We propose deregulation of vitamin A metabolism as a risk factor for benign and malignant prostate disease, and identified cancer associated FOXA1 indels affecting residue F254 as loss-of-function mutations promoting dedifferentiation of adult prostate progenitors. Summary: Retinoic acid signaling orchestrates luminal differentiation of adult prostate progenitors.
Collapse
|
21
|
Piazza A, Carlone R, Spencer GE. Non-canonical retinoid signaling in neural development, regeneration and synaptic function. Front Mol Neurosci 2024; 17:1371135. [PMID: 38516042 PMCID: PMC10954794 DOI: 10.3389/fnmol.2024.1371135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Canonical retinoid signaling via nuclear receptors and gene regulation is critical for the initiation of developmental processes such as cellular differentiation, patterning and neurite outgrowth, but also mediates nerve regeneration and synaptic functions in adult nervous systems. In addition to canonical transcriptional regulation, retinoids also exert rapid effects, and there are now multiple lines of evidence supporting non-canonical retinoid actions outside of the nucleus, including in dendrites and axons. Together, canonical and non-canonical retinoid signaling provide the precise temporal and spatial control necessary to achieve the fine cellular coordination required for proper nervous system function. Here, we examine and discuss the evidence supporting non-canonical actions of retinoids in neural development and regeneration as well as synaptic function, including a review of the proposed molecular mechanisms involved.
Collapse
Affiliation(s)
| | | | - Gaynor E. Spencer
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
22
|
Li Z, Zheng D, Zhang T, Ruan S, Li N, Yu Y, Peng Y, Wang D. The roles of nuclear receptors in cholesterol metabolism and reverse cholesterol transport in nonalcoholic fatty liver disease. Hepatol Commun 2024; 8:e0343. [PMID: 38099854 PMCID: PMC10727660 DOI: 10.1097/hc9.0000000000000343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023] Open
Abstract
As the most prevalent chronic liver disease globally, NAFLD encompasses a pathological process that ranges from simple steatosis to NASH, fibrosis, cirrhosis, and HCC, closely associated with numerous extrahepatic diseases. While the initial etiology was believed to be hepatocyte injury caused by lipid toxicity from accumulated triglycerides, recent studies suggest that an imbalance of cholesterol homeostasis is of greater significance. The role of nuclear receptors in regulating liver cholesterol homeostasis has been demonstrated to be crucial. This review summarizes the roles and regulatory mechanisms of nuclear receptors in the 3 main aspects of cholesterol production, excretion, and storage in the liver, as well as their cross talk in reverse cholesterol transport. It is hoped that this review will offer new insights and theoretical foundations for the study of the pathogenesis and progression of NAFLD and provide new research directions for extrahepatic diseases associated with NAFLD.
Collapse
|
23
|
Bohn T, de Lera AR, Landrier JF, Rühl R. Carotenoid metabolites, their tissue and blood concentrations in humans and further bioactivity via retinoid receptor-mediated signalling. Nutr Res Rev 2023; 36:498-511. [PMID: 36380523 DOI: 10.1017/s095442242200021x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Many epidemiological studies have emphasised the relation between carotenoid dietary intake and their circulating concentrations and beneficial health effects, such as lower risk of cardiometabolic diseases and cancer. However, there is dispute as to whether the attributed health benefits are due to native carotenoids or whether they are instead induced by their metabolites. Several categories of metabolites have been reported, most notably involving (a) modifications at the cyclohexenyl ring or the polyene chain, such as epoxides and geometric isomers, (b) excentric cleavage metabolites with alcohol-, aldehyde- or carboxylic acid-functional groups or (c) centric cleaved metabolites with additional hydroxyl, aldehyde or carboxyl functionalities, not counting their potential phase-II glucuronidated / sulphated derivatives. Of special interest are the apo-carotenoids, which originate in the intestine and other tissues from carotenoid cleavage by β-carotene oxygenases 1/2 in a symmetrical / non-symmetrical fashion. These are more water soluble and more electrophilic and, therefore, putative candidates for interactions with transcription factors such as NF-kB and Nrf2, as well as ligands for RAR-RXR nuclear receptor interactions. In this review, we discuss in vivo detected apo-carotenoids, their reported tissue concentrations, and potential associated health effects, focusing exclusively on the human situation and based on quantified / semi-quantified carotenoid metabolites proven to be present in humans.
Collapse
Affiliation(s)
- Torsten Bohn
- Nutrition and Health Research Group, Precision Health Department, Luxembourg Institute of Health, 1 A-B, rue Thomas Edison, L-1445, Strassen, Luxembourg
| | - Angel R de Lera
- Departmento de Química Orgánica, Facultade de Química, CINBIO and IBIV, Universidade de Vigo, 36310 Vigo, Spain
| | | | - Ralph Rühl
- CISCAREX UG, Berlin, Germany
- Paprika Bioanalytics BT, Debrecen, Hungary
| |
Collapse
|
24
|
Leal AS, Hung PY, Chowdhury AS, Liby KT. Retinoid X Receptor agonists as selective modulators of the immune system for the treatment of cancer. Pharmacol Ther 2023; 252:108561. [PMID: 37952906 PMCID: PMC10704405 DOI: 10.1016/j.pharmthera.2023.108561] [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: 05/01/2023] [Revised: 09/28/2023] [Accepted: 11/06/2023] [Indexed: 11/14/2023]
Abstract
Upon heterodimerizing with other nuclear receptors, retinoid X receptors (RXR) act as ligand-dependent transcription factors, regulating transcription of critical signaling pathways that impact numerous hallmarks of cancer. By controlling both inflammation and immune responses, ligands that activate RXR can modulate the tumor microenvironment. Several small molecule agonists of these essential receptors have been synthesized. Historically, RXR agonists were tested for inhibition of growth in cancer cells, but more recent drug discovery programs screen new molecules for inhibition of inflammation or activation of immune cells. Bexarotene is the first successful example of an effective therapeutic that molecularly targets RXR; this drug was approved to treat cutaneous T cell lymphoma and is still used as a standard of care treatment for this disease. No additional RXR agonists have yet achieved FDA approval, but several promising novel compounds are being developed. In this review, we provide an overview of the multiple mechanisms by which RXR signaling regulates inflammation and tumor immunity. We also discuss the potential of RXR-dependent immune cell modulation for the treatment or prevention of cancer and concomitant challenges and opportunities.
Collapse
Affiliation(s)
- Ana S Leal
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States of America; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Pei-Yu Hung
- Department of Physiology, Michigan State University, East Lansing, MI, United States of America
| | - Afrin Sultana Chowdhury
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Karen T Liby
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States of America; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America.
| |
Collapse
|
25
|
Almaguer J, Hindle A, Lawrence JJ. The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer's Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue. Antioxidants (Basel) 2023; 12:1921. [PMID: 38001775 PMCID: PMC10669734 DOI: 10.3390/antiox12111921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 11/26/2023] Open
Abstract
There is accumulating evidence that vitamin A (VA) deficiency contributes to the pathogenesis and progression of Alzheimer's disease (AD). All-trans retinoic acid (ATRA), a metabolite of VA in the brain, serves distinct roles in the human hippocampus. Agonists of retinoic acid receptors (RAR), including ATRA, promote activation of the non-amyloidogenic pathway by enhancing expression of α-secretases, providing a mechanistic basis for delaying/preventing amyloid beta (Aβ) toxicity. However, whether ATRA is actually deficient in the hippocampi of patients with AD is not clear. Here, using a publicly available human transcriptomic dataset, we evaluated the extent to which ATRA-sensitive genes are dysregulated in hippocampal tissue from post-mortem AD brains, relative to age-matched controls. Consistent with ATRA deficiency, we found significant dysregulation of many ATRA-sensitive genes and significant upregulation of RAR co-repressors, supporting the idea of transcriptional repression of ATRA-mediated signaling. Consistent with oxidative stress and neuroinflammation, Nrf2 and NfkB transcripts were upregulated, respectively. Interestingly, transcriptional targets of Nrf2 were not upregulated, accompanied by upregulation of several histone deacetylases. Overall, our investigation of ATRA-sensitive genes in the human hippocampus bolsters the scientific premise of ATRA depletion in AD and that epigenetic factors should be considered and addressed as part of VA supplementation.
Collapse
Affiliation(s)
- Joey Almaguer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Ashly Hindle
- Department of Pharmacology and Neuroscience and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - J. Josh Lawrence
- Department of Pharmacology and Neuroscience, Garrison Institute on Aging, Center of Excellence for Translational Neuroscience and Therapeutics, and Center of Excellence for Integrated Health, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
26
|
Yoo HS, Moss KO, Cockrum MA, Woo W, Napoli JL. Energy status regulates levels of the RAR/RXR ligand 9-cis-retinoic acid in mammalian tissues: Glucose reduces its synthesis in β-cells. J Biol Chem 2023; 299:105255. [PMID: 37714463 PMCID: PMC10582780 DOI: 10.1016/j.jbc.2023.105255] [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: 08/05/2023] [Revised: 09/01/2023] [Accepted: 09/10/2023] [Indexed: 09/17/2023] Open
Abstract
9-cis-retinoic acid (9cRA) binds retinoic acid receptors (RAR) and retinoid X receptors (RXR) with nanomolar affinities, in contrast to all-trans-retinoic acid (atRA), which binds only RAR with nanomolar affinities. RXR heterodimerize with type II nuclear receptors, including RAR, to regulate a vast gene array. Despite much effort, 9cRA has not been identified as an endogenous retinoid, other than in pancreas. By revising tissue analysis methods, 9cRA quantification by liquid chromatography-tandem mass spectrometry becomes possible in all mouse tissues analyzed. 9cRA occurs in concentrations similar to or greater than atRA. Fasting increases 9cRA in white and brown adipose, brain and pancreas, while increasing atRA in white adipose, liver and pancreas. 9cRA supports FoxO1 actions in pancreas β-cells and counteracts glucose actions that lead to glucotoxicity; in part by inducing Atg7 mRNA, which encodes the key enzyme essential for autophagy. Glucose suppresses 9cRA biosynthesis in the β-cell lines 832/13 and MIN6. Glucose reduces 9cRA biosynthesis in 832/13 cells by inhibiting Rdh5 transcription, unconnected to insulin, through cAMP and Akt, and inhibiting FoxO1. Through adapting tissue specifically to fasting, 9cRA would act independent of atRA. Widespread occurrence of 9cRA in vivo, and its self-sufficient adaptation to energy status, provides new perspectives into regulation of energy balance, attenuation of insulin and glucose actions, regulation of type II nuclear receptors, and retinoid biology.
Collapse
Affiliation(s)
- Hong Sik Yoo
- Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, Berkeley, California, USA
| | - Kristin Obrochta Moss
- Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, Berkeley, California, USA
| | - Michael A Cockrum
- Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, Berkeley, California, USA
| | - Wonsik Woo
- Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, Berkeley, California, USA
| | - Joseph L Napoli
- Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, Berkeley, California, USA.
| |
Collapse
|
27
|
Ito-Harashima S, Tsubouchi Y, Takada E, Kawanishi M, Yagi T. Development of a yeast reporter gene assay to detect ligands of freshwater cladoceran Daphnia magna ultraspiracle, a homolog of vertebrate retinoid X receptors. J Appl Toxicol 2023; 43:1447-1461. [PMID: 37078133 DOI: 10.1002/jat.4476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Endocrine-disrupting chemicals (EDCs) often affect homeostatic regulation in living organisms by directly acting on nuclear receptors (NRs). Retinoid X receptors (RXRs), the most highly conserved members of the NR superfamily during evolution, function as partners to form heterodimers with other NRs, such as retinoic acid, thyroid hormone, and vitamin D3 receptors. RXRs also homodimerize and induce the expression of target genes upon binding with their natural ligand, 9-cis-retinoic acid (9cRA), and typical EDCs organotin compounds, such as tributyltin and triphenyltin. In the present study, we established a new yeast reporter gene assay (RGA) to detect the ligands of freshwater cladoceran Daphnia magna ultraspiracle (Dapma-USP), a homolog of vertebrate RXRs. D. magna has been used as a representative crustacean species for aquatic EDC assessments in the Organization for Economic Corporation and Development test guidelines. Dapma-USP was expressed along with the Drosophila melanogaster steroid receptor coactivator Taiman in yeast cells carrying the lacZ reporter plasmid. The RGA for detecting agonist activity of organotins and o-butylphenol was improved by use of mutant yeast strains lacking genes encoding cell wall mannoproteins and/or plasma membrane drug efflux pumps as hosts. We also showed that a number of other human RXR ligands, phenol and bisphenol A derivatives, and terpenoid compounds such as 9c-RA exhibited antagonist activity on Dapma-USP. Our newly established yeast-based RGA system is valuable as the first screening tool to detect ligand substances for Dapma-USP and for evaluating the evolutionary divergence of the ligand responses of RXR homologs between humans and D. magna.
Collapse
Affiliation(s)
- Sayoko Ito-Harashima
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Japan
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Japan
| | - Yumiko Tsubouchi
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Japan
| | - Eiji Takada
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Japan
| | - Masanobu Kawanishi
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Japan
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Sakai, Japan
| | - Takashi Yagi
- Department of Biological Science, Graduate School of Science, Osaka Prefecture University, Sakai, Japan
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, Sakai, Japan
| |
Collapse
|
28
|
Wong NK, Yip SP, Huang CL. Establishing Functional Retina in a Dish: Progress and Promises of Induced Pluripotent Stem Cell-Based Retinal Neuron Differentiation. Int J Mol Sci 2023; 24:13652. [PMID: 37686457 PMCID: PMC10487913 DOI: 10.3390/ijms241713652] [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: 08/03/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
The human eye plays a critical role in vision perception, but various retinal degenerative diseases such as retinitis pigmentosa (RP), glaucoma, and age-related macular degeneration (AMD) can lead to vision loss or blindness. Although progress has been made in understanding retinal development and in clinical research, current treatments remain inadequate for curing or reversing these degenerative conditions. Animal models have limited relevance to humans, and obtaining human eye tissue samples is challenging due to ethical and legal considerations. Consequently, researchers have turned to stem cell-based approaches, specifically induced pluripotent stem cells (iPSCs), to generate distinct retinal cell populations and develop cell replacement therapies. iPSCs offer a novel platform for studying the key stages of human retinogenesis and disease-specific mechanisms. Stem cell technology has facilitated the production of diverse retinal cell types, including retinal ganglion cells (RGCs) and photoreceptors, and the development of retinal organoids has emerged as a valuable in vitro tool for investigating retinal neuron differentiation and modeling retinal diseases. This review focuses on the protocols, culture conditions, and techniques employed in differentiating retinal neurons from iPSCs. Furthermore, it emphasizes the significance of molecular and functional validation of the differentiated cells.
Collapse
Affiliation(s)
- Nonthaphat Kent Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| | - Chien-Ling Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| |
Collapse
|
29
|
Liu Y, Wang P, Jin G, Shi P, Zhao Y, Guo J, Yin Y, Shao Q, Li P, Yang P. The novel function of bexarotene for neurological diseases. Ageing Res Rev 2023; 90:102021. [PMID: 37495118 DOI: 10.1016/j.arr.2023.102021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023]
Abstract
Bexarotene, a retinoid X receptor (RXR) agonist, is approved by FDA to treat cutaneous T-cell lymphoma. However, it has also demonstrated promising therapeutic potential for neurological diseases such as stroke, traumatic brain injury, Parkinson's disease, and particularly Alzheimer's disease(AD). In AD, bexarotene inhibits the production and aggregation of amyloid β (Aβ), activates Liver X Receptor/RXR heterodimers to increase lipidated apolipoprotein E to remove Aβ, mitigates the negative impact of Aβ, regulates neuroinflammation, and ultimately improves cognitive function. For other neurological diseases, its mechanisms of action include inhibiting inflammatory responses, up-regulating microglial phagocytosis, and reducing misfolded protein aggregation, all of which aid in alleviating neurological damage. Here, we briefly discuss the characteristics, applications, and adverse effects of bexarotene, summarize its pharmacological mechanisms and therapeutic results in various neurological diseases, and elaborate on the problems encountered in preclinical research, with the aim of providing help for the further application of bexarotene in central nervous system diseases.
Collapse
Affiliation(s)
- Yangtao Liu
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; College of Third Clinical, Xinxiang Medical University, Xinxiang, China
| | - Pengwei Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Weihui, China
| | - Guofang Jin
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Peijie Shi
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yonghui Zhao
- Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiayi Guo
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Yaling Yin
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Qianhang Shao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China.
| | - Peng Li
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| | - Pengfei Yang
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| |
Collapse
|
30
|
Manoharan I, Shanmugam A, Ramalingam M, Patel N, Thangaraju M, Ande S, Pacholczyk R, Prasad PD, Manicassamy S. The Transcription Factor RXRα in CD11c+ APCs Regulates Intestinal Immune Homeostasis and Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:853-861. [PMID: 37477694 PMCID: PMC10538854 DOI: 10.4049/jimmunol.2200909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
APCs such as dendritic cells and macrophages play a pivotal role in mediating immune tolerance and restoring intestinal immune homeostasis by limiting inflammatory responses against commensal bacteria. However, cell-intrinsic molecular regulators critical for programming intestinal APCs to a regulatory state rather than an inflammatory state are unknown. In this study, we report that the transcription factor retinoid X receptor α (RXRα) signaling in CD11c+ APCs is essential for suppressing intestinal inflammation by imparting an anti-inflammatory phenotype. Using a mouse model of ulcerative colitis, we demonstrated that targeted deletion of RXRα in CD11c+ APCs in mice resulted in the loss of T cell homeostasis with enhanced intestinal inflammation and increased histopathological severity of colonic tissue. This was due to the increased production of proinflammatory cytokines that drive Th1/Th17 responses and decreased expression of immune-regulatory factors that promote regulatory T cell differentiation in the colon. Consistent with these findings, pharmacological activation of the RXRα pathway alleviated colitis severity in mice by suppressing the expression of inflammatory cytokines and limiting Th1/Th17 cell differentiation. These findings identify an essential role for RXRα in APCs in regulating intestinal immune homeostasis and inflammation. Thus, manipulating the RXRα pathway could provide novel opportunities for enhancing regulatory responses and dampening colonic inflammation.
Collapse
Affiliation(s)
- Indumathi Manoharan
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | | | | | - Nikhil Patel
- Department of Pathology, Augusta University, Augusta, GA USA
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Satyanarayana Ande
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | | | - Puttur D. Prasad
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Santhakumar Manicassamy
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
- Department of Medicine, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
31
|
Bohn T, Hellman-Regen J, de Lera AR, Böhm V, Rühl R. Human nutritional relevance and suggested nutritional guidelines for vitamin A5/X and provitamin A5/X. Nutr Metab (Lond) 2023; 20:34. [PMID: 37582723 PMCID: PMC10426203 DOI: 10.1186/s12986-023-00750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 05/27/2023] [Indexed: 08/17/2023] Open
Abstract
In the last century, vitamin A was identified that included the nutritional relevant vitamin A1 / provitamin A1, as well as the vitamin A2 pathway concept. Globally, nutritional guidelines have focused on vitamin A1 with simplified recommendations and calculations based solely on vitamin A. The vitamin A / provitamin A terminology described vitamin A with respect to acting as a precursor of 11-cis-retinal, the chromophore of the visual pigment, as well as retinoic acid(s), being ligand(s) of the nuclear hormone receptors retinoic acid receptors (RARs) α, β and γ. All-trans-retinoic acid was conclusively shown to be the endogenous RAR ligand, while the concept of its isomer 9-cis-retinoic acid, being "the" endogenous ligand of the retinoid-X receptors (RXRs), remained inconclusive. Recently, 9-cis-13,14-dihydroretinoic acid was conclusively reported as an endogenous RXR ligand, and a direct nutritional precursor was postulated in 2018 and further confirmed by Rühl, Krezel and de Lera in 2021. This was further termed vitamin A5/X / provitamin A5/X. In this review, a new vitamin A5/X / provitamin A5/X concept is conceptualized in parallel to the vitamin A(1) / provitamin A(1) concept for daily dietary intake and towards dietary guidelines, with a focus on the existing national and international regulations for the physiological and nutritional relevance of vitamin A5/X. The aim of this review is to summarize available evidence and to emphasize gaps of knowledge regarding vitamin A5/X, based on new and older studies and proposed future directions as well as to stimulate and propose adapted nutritional regulations.
Collapse
Affiliation(s)
- Torsten Bohn
- Nutrition Research Group, Department of Precision Health, Luxembourg Institute and Health, 1 A-B, Rue Thomas Edison, 1445, Strassen, Luxembourg
| | - Julian Hellman-Regen
- Department of Psychiatry, Charité-Campus Benjamin Franklin, Section Neurobiology, University Medicine Berlin, Berlin, Germany
| | - Angel R de Lera
- Departamento de Química Orgánica, Facultad de Química, CINBIO and IBIV, Universidade de Vigo, Campus As Lagoas-Marcosende, 36310, Vigo, Spain
| | - Volker Böhm
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Ralph Rühl
- CISCAREX UG, Transvaalstr. 27c, 13351, Berlin, Germany.
| |
Collapse
|
32
|
Chen J, Zhao T, He F, Zhong Y, Wang S, Tang Z, Qiu Y, Wu Z, Fang M. Discovery of bipyridine amide derivatives targeting pRXRα-PLK1 interaction for anticancer therapy. Eur J Med Chem 2023; 254:115341. [PMID: 37058970 DOI: 10.1016/j.ejmech.2023.115341] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/23/2023] [Accepted: 04/02/2023] [Indexed: 04/16/2023]
Abstract
Retinoid X receptor alpha (RXRα) is an important therapeutic target of cancer. Recently, small molecules (e.g.,XS-060 and its derivatives), which can significantly induce RXRα-dependent mitotic arrest by inhibiting pRXRα-PLK1 interaction, have been demonstrated as excellent anticancer agents. To further obtain novel RXR-targeted antimitotic agents with excellent bioactivity and drug-like properties, we herein synthesized two new series of bipyridine amide derivatives with XS-060 as the lead compound. In the reporter gene assay, most synthesized compounds showed antagonistic activity against RXRα. The most active compound, bipyridine amide B9 (BPA-B9), showed better activity than XS-060, with excellent RXRα-binding affinity (KD = 39.29 ± 1.12 nM) and anti-proliferative activity against MDA-MB-231 (IC50 = 16 nM, SI > 3). Besides, a docking study revealed a proper fitting of BPA-B9 into the coactivator binding site of RXRα, rationalizing its potent antagonistic effect on RXRα transactivation. Further, the mechanism studies revealed that the anticancer activity of BPA-B9 was dependent on its cellular RXRα-targeted mechanism, such as inhibiting pRXRα-PLK1 interaction and inducing RXRα-dependent mitotic arrest. Besides, BPA-B9 displayed better pharmacokinetics than the lead XS-060. Further, animal assays indicated BPA-B9 had significant anticancer efficacy in vivo with no considerable side effects. Together, our study reveals a novel RXRα ligand BPA-B9 targeting the pRXRα-PLK1 interaction, with great potential as a promising anticancer drug candidate for further development.
Collapse
Affiliation(s)
- Jun Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Taige Zhao
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Fengming He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Yijing Zhong
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Susu Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Ziqing Tang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Yingkun Qiu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China
| | - Zhen Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China.
| | - Meijuan Fang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Science, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
33
|
Zhou P, Pu WT. Molecule in mothers' milk nurses pups' heart cells to maturity. Nature 2023; 618:242-243. [PMID: 37225793 DOI: 10.1038/d41586-023-01635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
|
34
|
Stewart KS, Gonzales KAU, Yuan S, Tierney MT, Bonny AR, Yang Y, Infarinato NR, Cowley CJ, Levorse JM, Pasolli HA, Ghosh S, Rothlin CV, Fuchs E. Stem cells tightly regulate dead cell clearance to maintain tissue fitness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541773. [PMID: 37293114 PMCID: PMC10245816 DOI: 10.1101/2023.05.22.541773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Macrophages and dendritic cells have long been appreciated for their ability to migrate to and engulf dying cells and debris, including some of the billions of cells that are naturally eliminated from our body daily. However, a substantial number of these dying cells are cleared by 'non-professional phagocytes', local epithelial cells that are critical to organismal fitness. How non-professional phagocytes sense and digest nearby apoptotic corpses while still performing their normal tissue functions is unclear. Here, we explore the molecular mechanisms underlying their multifunctionality. Exploiting the cyclical bouts of tissue regeneration and degeneration during the hair cycle, we show that stem cells can transiently become non-professional phagocytes when confronted with dying cells. Adoption of this phagocytic state requires both local lipids produced by apoptotic corpses to activate RXRα, and tissue-specific retinoids for RARγ activation. This dual factor dependency enables tight regulation of the genes requisite to activate phagocytic apoptotic clearance. The tunable phagocytic program we describe here offers an effective mechanism to offset phagocytic duties against the primary stem cell function of replenishing differentiated cells to preserve tissue integrity during homeostasis. Our findings have broad implications for other non-motile stem or progenitor cells which experience cell death in an immune-privileged niche.
Collapse
Affiliation(s)
- Katherine S Stewart
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Kevin AU Gonzales
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Shaopeng Yuan
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Matthew T Tierney
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Alain R Bonny
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Yihao Yang
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Nicole R Infarinato
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Christopher J Cowley
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - John M Levorse
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Hilda Amalia Pasolli
- Electron Microscopy Resource Center, The Rockefeller University, New York, NY, USA
| | - Sourav Ghosh
- Departments of Neurology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Carla V Rothlin
- Departments of Immunobiology and Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| |
Collapse
|
35
|
Melo N, Belyaeva OV, Berger WK, Halasz L, Yu J, Pilli N, Yang Z, Klyuyeva AV, Elmets CA, Atigadda V, Muccio DD, Kane MA, Nagy L, Kedishvili NY, Renfrow MB. Next-generation retinoid X receptor agonists increase ATRA signaling in organotypic epithelium cultures and have distinct effects on receptor dynamics. J Biol Chem 2023; 299:102746. [PMID: 36436565 PMCID: PMC9807999 DOI: 10.1016/j.jbc.2022.102746] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/26/2022] Open
Abstract
Retinoid X receptors (RXRs) are nuclear transcription factors that partner with other nuclear receptors to regulate numerous physiological processes. Although RXR represents a valid therapeutic target, only a few RXR-specific ligands (rexinoids) have been identified, in part due to the lack of clarity on how rexinoids selectively modulate RXR response. Previously, we showed that rexinoid UAB30 potentiates all-trans-retinoic acid (ATRA) signaling in human keratinocytes, in part by stimulating ATRA biosynthesis. Here, we examined the mechanism of action of next-generation rexinoids UAB110 and UAB111 that are more potent in vitro than UAB30 and the FDA-approved Targretin. Both UAB110 and UAB111 enhanced ATRA signaling in human organotypic epithelium at a 50-fold lower concentration than UAB30. This was consistent with the 2- to 5- fold greater increase in ATRA in organotypic epidermis treated with UAB110/111 versus UAB30. Furthermore, at 0.2 μM, UAB110/111 increased the expression of ATRA genes up to 16-fold stronger than Targretin. The less toxic and more potent UAB110 also induced more changes in differential gene expression than Targretin. Additionally, our hydrogen deuterium exchange mass spectrometry analysis showed that both ligands reduced the dynamics of the ligand-binding pocket but also induced unique dynamic responses that were indicative of higher affinity binding relative to UAB30, especially for Helix 3. UAB110 binding also showed increased dynamics towards the dimer interface through the Helix 8 and Helix 9 regions. These data suggest that UAB110 and UAB111 are potent activators of RXR-RAR signaling pathways but accomplish activation through different molecular responses to ligand binding.
Collapse
Affiliation(s)
- Nathalia Melo
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Olga V Belyaeva
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wilhelm K Berger
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Laszlo Halasz
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland, USA
| | - Nagesh Pilli
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland, USA
| | - Zhengrong Yang
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alla V Klyuyeva
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Craig A Elmets
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Venkatram Atigadda
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Donald D Muccio
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland, USA
| | - Laszlo Nagy
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Natalia Y Kedishvili
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | - Matthew B Renfrow
- O'Neil Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
36
|
Abstract
For almost a century, vitamin A has been known as a nutrient critical for normal development, differentiation, and homeostasis; accordingly, there has been much interest in understanding its mechanism of action. This review is about the discovery of specific receptors for the vitamin A derivative, retinoic acid (RA), which launched extensive molecular, genetic, and structural investigations into these new members of the nuclear receptor superfamily of transcriptional regulators. These included two families of receptors, the RAR isotypes (α, β, and γ) along with three RXR isotypes (α, β, and γ), which bind as RXR/RAR heterodimers to cis-acting response elements of RA target genes to generate a high degree of complexity. Such studies have provided deep molecular insight into how the widespread pleiotropic effects of RA can be generated.
Collapse
Affiliation(s)
- Martin Petkovich
- Department of Pathology and Molecular Medicine, Queens University, Kingston, Ontario, Canada
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (I.G.B.M.C.), Illkirch, France
| |
Collapse
|
37
|
Rastinejad F. Retinoic acid receptor structures: the journey from single domains to full-length complex. J Mol Endocrinol 2022; 69:T25-T36. [PMID: 36069789 PMCID: PMC11376212 DOI: 10.1530/jme-22-0113] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/07/2022] [Indexed: 11/08/2022]
Abstract
The retinoic acid receptors (RARα, β, and γ) are multi-domain polypeptides that heterodimerize with retinoid X receptors (RXRα, β, and γ) to form functional transcription factors. Understanding the three-dimensional molecular organization of these nuclear receptors (NRs) began with RAR and RXR DNA-binding domains (DBDs), and were followed with studies on isolated ligand-binding domains (LBDs). The more complete picture emerged in 2017 with the multi-domain crystal structure of RXRα-RARβ on its response element with retinoic acid molecules and coactivator segments on both proteins. The analysis of that structure and its complementary studies have clarified the direct communication pathways within RXR-RAR polypeptides, through which DNA binding, protein-ligand, and protein-protein interactions are integrated for overall functional responses. Understanding the molecular connections in the RXR-RAR complex has benefited from direct observations of the multi-domain structures of RXRα-PPARγ, RXRα-LXRβ, HNF-4α homodimer, and androgen receptor homodimer, each bound to its response element. These comprehensive NR structures show unique quaternary architectures, yet all have DBD-DBD, LBD-LBD, and DBD-LBD domain-domain contacts within them. These convergence zones allow signals from discrete domains of their polypeptides to be propagated and integrated across their entire complex, shaping their overall responses in an allosteric fashion.
Collapse
Affiliation(s)
- Fraydoon Rastinejad
- Nuffield Department of Medicine, University of Oxford, Target Discovery Institute (NDM RB), Oxford, UK
| |
Collapse
|
38
|
Giguère V, Evans RM. Chronicle of a discovery: the retinoic acid receptor. J Mol Endocrinol 2022; 69:T1-T11. [PMID: 35900848 DOI: 10.1530/jme-22-0117] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 06/18/2022] [Indexed: 11/08/2022]
Abstract
The landmark 1987 discovery of the retinoic acid receptor (RAR) came as a surprise, uncovering a genomic kinship between the fields of vitamin A biology and steroid receptors. This stunning breakthrough triggered a cascade of studies to deconstruct the roles played by the RAR and its natural and synthetic ligands in embryonic development, skin, growth, physiology, vision, and disease as well as providing a template to elucidate the molecular mechanisms by which nuclear receptors regulate gene expression. In this review, written from historic and personal perspectives, we highlight the milestones that led to the discovery of the RAR and the subsequent studies that enriched our knowledge of the molecular mechanisms by which a low-abundant dietary compound could be so essential to the generation and maintenance of life itself.
Collapse
Affiliation(s)
- Vincent Giguère
- Goodman Cancer Institute, McGill University, Montréal, Quebec, Canada
- Department of Biochemistry, McGill University, Montréal, Quebec, Canada
| | - Ronald M Evans
- The Salk Institute for Biological Studies, La Jolla, California, USA
| |
Collapse
|
39
|
Arthur P, Muok L, Nathani A, Zeng EZ, Sun L, Li Y, Singh M. Bioengineering Human Pluripotent Stem Cell-Derived Retinal Organoids and Optic Vesicle-Containing Brain Organoids for Ocular Diseases. Cells 2022; 11:3429. [PMID: 36359825 PMCID: PMC9653705 DOI: 10.3390/cells11213429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 08/24/2023] Open
Abstract
Retinal organoids are three-dimensional (3D) structures derived from human pluripotent stem cells (hPSCs) that mimic the retina's spatial and temporal differentiation, making them useful as in vitro retinal development models. Retinal organoids can be assembled with brain organoids, the 3D self-assembled aggregates derived from hPSCs containing different cell types and cytoarchitectures that resemble the human embryonic brain. Recent studies have shown the development of optic cups in brain organoids. The cellular components of a developing optic vesicle-containing organoids include primitive corneal epithelial and lens-like cells, retinal pigment epithelia, retinal progenitor cells, axon-like projections, and electrically active neuronal networks. The importance of retinal organoids in ocular diseases such as age-related macular degeneration, Stargardt disease, retinitis pigmentosa, and diabetic retinopathy are described in this review. This review highlights current developments in retinal organoid techniques, and their applications in ocular conditions such as disease modeling, gene therapy, drug screening and development. In addition, recent advancements in utilizing extracellular vesicles secreted by retinal organoids for ocular disease treatments are summarized.
Collapse
Affiliation(s)
- Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Laureana Muok
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Eric Z. Zeng
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
40
|
le Maire A, Rey M, Vivat V, Guée L, Blanc P, Malosse C, Chamot-Rooke J, Germain P, Bourguet W. Design and in vitro characterization of RXR variants as tools to investigate the biological role of endogenous rexinoids. J Mol Endocrinol 2022; 69:377-390. [PMID: 35900852 DOI: 10.1530/jme-22-0021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 11/08/2022]
Abstract
Retinoid X receptors (RXRα, β, and γ) are essential members of the nuclear receptor (NR) superfamily of ligand-dependent transcriptional regulators that bind DNA response elements and control the expression of large gene networks. As obligate heterodimerization partners of many NRs, RXRs are involved in a variety of pathophysiological processes. However, despite this central role in NR signaling, there is still no consensus regarding the precise biological functions of RXRs and the putative role of the endogenous ligands (rexinoids) previously proposed for these receptors. Based on available crystal structures, we introduced a series of amino acid substitutions into the ligand-binding pocket of all three RXR subtypes in order to alter their binding properties. Subsequent characterization using a battery of cell-based and in vitro assays led to the identification of a double mutation abolishing the binding of any ligand while keeping the other receptor functions intact and a triple mutation that selectively impairs interaction with natural rexinoids but not with some synthetic ligands. We also report crystal structures that help understand the specific ligand-binding capabilities of both variants. These RXR variants, either fully disabled for ligand binding or retaining the property of being activated by synthetic compounds, represent unique tools that could be used in future studies to probe the presence of active endogenous rexinoids in tissues/organs and to investigate their role in vivo. Last, we provide data suggesting a possible involvement of fatty acids in the weak interaction of RXRs with corepressors.
Collapse
Affiliation(s)
- Albane le Maire
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Martial Rey
- Institut Pasteur, Université de Paris, CNRS USR2000, Mass Spectrometry for Biology Unit, Paris, France
| | - Valérie Vivat
- IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Univ Strasbourg, CNRS, Inserm, Illkirch, France
| | - Laura Guée
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Pauline Blanc
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - Christian Malosse
- Institut Pasteur, Université de Paris, CNRS USR2000, Mass Spectrometry for Biology Unit, Paris, France
| | - Julia Chamot-Rooke
- Institut Pasteur, Université de Paris, CNRS USR2000, Mass Spectrometry for Biology Unit, Paris, France
| | - Pierre Germain
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| | - William Bourguet
- CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS, Inserm, Montpellier, France
| |
Collapse
|
41
|
de Hoog E, Saba Echezarreta VE, Turgambayeva A, Foran G, Megaly M, Necakov A, Spencer GE. Molluscan RXR Transcriptional Regulation by Retinoids in a Drosophila CNS Organ Culture System. Cells 2022; 11:2493. [PMID: 36010570 PMCID: PMC9406730 DOI: 10.3390/cells11162493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/26/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022] Open
Abstract
Retinoic acid, the active metabolite of Vitamin A, is important for the appropriate development of the nervous system (e.g., neurite outgrowth) as well as for cognition (e.g., memory formation) in the adult brain. We have shown that many of the effects of retinoids are conserved in the CNS of the mollusc, Lymnaea stagnalis. RXRs are predominantly nuclear receptors, but the Lymnaea RXR (LymRXR) exhibits a non-nuclear distribution in the adult CNS, where it is also implicated in non-genomic retinoid functions. As such, we developed a CNS Drosophila organ culture-based system to examine the transcriptional activity and ligand-binding properties of LymRXR, in the context of a live invertebrate nervous system. The novel ligand sensor system was capable of reporting both the expression and transcriptional activity of the sensor. Our results indicate that the LymRXR ligand sensor mediated transcription following activation by both 9-cis RA (the high affinity ligand for vertebrate RXRs) as well as the vertebrate RXR synthetic agonist, SR11237. The LymRXR ligand sensor was also activated by all-trans RA, and to a much lesser extent by the vertebrate RAR synthetic agonist, EC23. This sensor also detected endogenous retinoid-like activity in the CNS of developing Drosophila larvae, primarily during the 3rd instar larval stage. These data indicate that the LymRXR sensor can be utilized not only for characterization of ligand activation for studies related to the Lymnaea CNS, but also for future studies of retinoids and their functions in Drosophila development.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gaynor E. Spencer
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
42
|
Deng Q, Chen J. Potential Therapeutic Effect of All-Trans Retinoic Acid on Atherosclerosis. Biomolecules 2022; 12:869. [PMID: 35883425 PMCID: PMC9312697 DOI: 10.3390/biom12070869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/08/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is a major risk factor for myocardial infarction and ischemic stroke, which are the leading cause of death worldwide. All-trans retinoic acid (ATRA) is a natural derivative of essential vitamin A. Numerous studies have shown that ATRA plays an important role in cell proliferation, cell apoptosis, cell differentiation, and embryonic development. All-trans retinoic acid (ATRA) is a ligand of retinoic acid receptors that regulates various biological processes by activating retinoic acid signals. In this paper, the metabolic processes of ATRA were reviewed, with emphasis on the effects of ATRA on inflammatory cells involved in the process of atherosclerosis.
Collapse
Affiliation(s)
| | - Jixiang Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
43
|
Tarnow G, McLachlan A. Selective effect of β-catenin on nuclear receptor-dependent hepatitis B virus transcription and replication. Virology 2022; 571:52-58. [PMID: 35509137 DOI: 10.1016/j.virol.2022.04.006] [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: 12/23/2021] [Revised: 03/07/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
β-catenin regulates HBV transcription in cell culture and viral biosynthesis in vivo in the transgenic mouse model of chronic HBV infection. Therefore, it is important to understand which transcription factor activities are coactivated by β-catenin to enhance HBV biosynthesis. The effect of β-catenin expression in the context of nuclear receptor-mediated HBV transcription was evaluated initially in the human embryonic kidney cell line, HEK293T. Reporter gene and viral replication assays revealed that β-catenin can coactivate HBV transcription through some, most predominantly liver receptor homolog 1 (LRH1), but not all nuclear receptors known to activate viral biosynthesis. Similarly, β-catenin activated nuclear receptor-mediated HBV transcription and replication in the human hepatoma cell line, Huh7, primarily through its effect on the farnesoid X receptor α (FXRα). These data indicate that β-catenin can enhance nuclear receptor-mediated HBV biosynthesis, but the relative importance of various transcription factors is dependent upon the precise cellular environment.
Collapse
Affiliation(s)
- Grant Tarnow
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 909 South Wolcott Avenue, Chicago, IL, 60612, USA
| | - Alan McLachlan
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 909 South Wolcott Avenue, Chicago, IL, 60612, USA.
| |
Collapse
|
44
|
Petermann AB, Reyna-Jeldes M, Ortega L, Coddou C, Yévenes GE. Roles of the Unsaturated Fatty Acid Docosahexaenoic Acid in the Central Nervous System: Molecular and Cellular Insights. Int J Mol Sci 2022; 23:5390. [PMID: 35628201 PMCID: PMC9141004 DOI: 10.3390/ijms23105390] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 11/16/2022] Open
Abstract
Fatty acids (FAs) are essential components of the central nervous system (CNS), where they exert multiple roles in health and disease. Among the FAs, docosahexaenoic acid (DHA) has been widely recognized as a key molecule for neuronal function and cell signaling. Despite its relevance, the molecular pathways underlying the beneficial effects of DHA on the cells of the CNS are still unclear. Here, we summarize and discuss the molecular mechanisms underlying the actions of DHA in neural cells with a special focus on processes of survival, morphological development, and synaptic maturation. In addition, we examine the evidence supporting a potential therapeutic role of DHA against CNS tumor diseases and tumorigenesis. The current results suggest that DHA exerts its actions on neural cells mainly through the modulation of signaling cascades involving the activation of diverse types of receptors. In addition, we found evidence connecting brain DHA and ω-3 PUFA levels with CNS diseases, such as depression, autism spectrum disorders, obesity, and neurodegenerative diseases. In the context of cancer, the existing data have shown that DHA exerts positive actions as a coadjuvant in antitumoral therapy. Although many questions in the field remain only partially resolved, we hope that future research may soon define specific pathways and receptor systems involved in the beneficial effects of DHA in cells of the CNS, opening new avenues for innovative therapeutic strategies for CNS diseases.
Collapse
Affiliation(s)
- Ana B. Petermann
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070386, Chile;
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
| | - Mauricio Reyna-Jeldes
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Lorena Ortega
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Claudio Coddou
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica Del Norte, Coquimbo 1781421, Chile
- Núcleo para el Estudio del Cáncer a Nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta 1270709, Chile
| | - Gonzalo E. Yévenes
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción 4070386, Chile;
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago 8330025, Chile; (M.R.-J.); (L.O.)
| |
Collapse
|
45
|
Pípal M, Novák J, Rafajová A, Smutná M, Hilscherová K. Teratogenicity of retinoids detected in surface waters in zebrafish embryos and its predictability by in vitro assays. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 246:106151. [PMID: 35390581 DOI: 10.1016/j.aquatox.2022.106151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/08/2022] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
Retinoids are newly detected compounds in aquatic ecosystems associated with cyanobacterial water blooms. Their potential health risks are only scarcely described despite numerous detections of all-trans retinoic acid (ATRA) and its derivatives in the environment. Besides the known teratogen ATRA there is only little or no information about their potency and namely their effects in vivo. We characterize ATRA and 8 other retinoids reported to occur in the environment for their bioactivity and teratogenicity using four in vitro reporter gene assays and zebrafish (Danio rerio) embryotoxicity assay. Our results document the ability of these compounds to interfere with retinoid signalling and cause teratogenicity at environmentally relevant levels with EC50 values at nM (hundreds of ng/L) levels and teratogenic indexes ranging from 2.8 (9cis retinoic acid) to 15.8 (retinal). The relative potency of individual compounds for teratogenicity ranged from 0.059 (retinal) to 0.96 (5,6-epoxy ATRA) when compared to ATRA. An environmentally relevant mixture of retinoids was tested showing good predictability of teratogenicity from the in vitro activities and additive toxicity of the mixture. The high teratogenicity of the newly described compounds associated with cyanobacteria presents a concern for developmental stages due to high conservation of the retinoid signalling across vertebrates.
Collapse
Affiliation(s)
- Marek Pípal
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Jiří Novák
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Aneta Rafajová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Marie Smutná
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic
| | - Klára Hilscherová
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Kamenice, Brno 62500 , Czech Republic.
| |
Collapse
|
46
|
Antagonizing RARγ Drives Necroptosis of Cancer Stem Cells. Int J Mol Sci 2022; 23:ijms23094814. [PMID: 35563205 PMCID: PMC9105400 DOI: 10.3390/ijms23094814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 12/23/2022] Open
Abstract
There is a need for agents that eliminate cancer stem cells, which sustain cancer and are also largely responsible for disease relapse and metastasis. Conventional chemotherapeutics and radiotherapy are often highly effective against the bulk of cancer cells, which are proliferating, but spare cancer stem cells. Therapeutics that target cancer stem cells may also provide a bona fide cure for cancer. There are two rationales for targeting the retinoic acid receptor (RAR)γ. First, RARγ is expressed selectively within primitive cells. Second, RARγ is a putative oncogene for a number of human cancers, including cases of acute myeloid leukemia, cholangiocarcinoma, and colorectal, renal and hepatocellular carcinomas. Prostate cancer cells depend on active RARγ for their survival. Antagonizing all RARs caused necroptosis of prostate and breast cancer stem cell-like cells, and the cancer stem cells that gave rise to neurospheres from pediatric patients’ primitive neuroectodermal tumors and an astrocytoma. As tested for prostate cancer, antagonizing RARγ was sufficient to drive necroptosis. Achieving cancer-selectively is a longstanding paradigm for developing new treatments. The normal prostate epithelium was less sensitive to the RARγ antagonist and pan-RAR antagonist than prostate cancer cells, and fibroblasts and blood mononuclear cells were insensitive. The RARγ antagonist and pan-RAR antagonist are promising new cancer therapeutics.
Collapse
|
47
|
Triple chemical derivatization strategy assisted liquid chromatography-mass spectrometry for determination of retinoic acids in human serum. Talanta 2022; 245:123474. [DOI: 10.1016/j.talanta.2022.123474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 11/18/2022]
|
48
|
O’Connor C, Varshosaz P, Moise AR. Mechanisms of Feedback Regulation of Vitamin A Metabolism. Nutrients 2022; 14:nu14061312. [PMID: 35334970 PMCID: PMC8950952 DOI: 10.3390/nu14061312] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Vitamin A is an essential nutrient required throughout life. Through its various metabolites, vitamin A sustains fetal development, immunity, vision, and the maintenance, regulation, and repair of adult tissues. Abnormal tissue levels of the vitamin A metabolite, retinoic acid, can result in detrimental effects which can include congenital defects, immune deficiencies, proliferative defects, and toxicity. For this reason, intricate feedback mechanisms have evolved to allow tissues to generate appropriate levels of active retinoid metabolites despite variations in the level and format, or in the absorption and conversion efficiency of dietary vitamin A precursors. Here, we review basic mechanisms that govern vitamin A signaling and metabolism, and we focus on retinoic acid-controlled feedback mechanisms that contribute to vitamin A homeostasis. Several approaches to investigate mechanistic details of the vitamin A homeostatic regulation using genomic, gene editing, and chromatin capture technologies are also discussed.
Collapse
Affiliation(s)
- Catherine O’Connor
- MD Program, Northern Ontario School of Medicine, 317-MSE Bldg., 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada;
| | - Parisa Varshosaz
- Biology and Biomolecular Sciences Ph.D. Program, Northern Ontario School of Medicine, Laurentian University, Sudbury, ON P3E 2C6, Canada;
| | - Alexander R. Moise
- Medical Sciences Division, Northern Ontario School of Medicine, 317-MSE Bldg., 935 Ramsey Lake Rd., Sudbury, ON P3E 2C6, Canada
- Department of Chemistry and Biochemistry, Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: ; Tel.: +1-705-662-7253
| |
Collapse
|
49
|
Steinhoff JS, Lass A, Schupp M. Retinoid Homeostasis and Beyond: How Retinol Binding Protein 4 Contributes to Health and Disease. Nutrients 2022; 14:1236. [PMID: 35334893 PMCID: PMC8951293 DOI: 10.3390/nu14061236] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
Retinol binding protein 4 (RBP4) is the specific transport protein of the lipophilic vitamin A, retinol, in blood. Circulating RBP4 originates from the liver. It is secreted by hepatocytes after it has been loaded with retinol and binding to transthyretin (TTR). TTR association prevents renal filtration due to the formation of a higher molecular weight complex. In the circulation, RBP4 binds to specific membrane receptors, thereby delivering retinol to target cells, rendering liver-secreted RBP4 the major mechanism to distribute hepatic vitamin A stores to extrahepatic tissues. In particular, binding of RBP4 to 'stimulated by retinoic acid 6' (STRA6) is required to balance tissue retinoid responses in a highly homeostatic manner. Consequently, defects/mutations in RBP4 can cause a variety of conditions and diseases due to dysregulated retinoid homeostasis and cover embryonic development, vision, metabolism, and cardiovascular diseases. Aside from the effects related to retinol transport, non-canonical functions of RBP4 have also been reported. In this review, we summarize the current knowledge on the regulation and function of RBP4 in health and disease derived from murine models and human mutations.
Collapse
Affiliation(s)
- Julia S. Steinhoff
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular Metabolic Renal (CMR)-Research Center, 10115 Berlin, Germany;
| | - Achim Lass
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Heinrichstraße 31/II, A-8010 Graz, Austria;
- Field of Excellence BioHealth, University of Graz, Heinrichstraße 31/II, A-8010 Graz, Austria
| | - Michael Schupp
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular Metabolic Renal (CMR)-Research Center, 10115 Berlin, Germany;
| |
Collapse
|
50
|
Zhao A, Miao J, Liu L, Pan L. Potencies of organotin compounds in scallop RXRa responsive activity with a GAL4-based reconstituted yeast assay in vitro. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:19890-19897. [PMID: 35084679 DOI: 10.1007/s11356-022-18620-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 01/07/2022] [Indexed: 06/14/2023]
Abstract
Retinoid X receptor (RXR) has been found to be a major target in various processes of endocrine disruption from the exposure to organotin compounds (OTCs), including imposex in gastropod mollusks. It was also reported in bivalves that OTCs caused intersex and skewed sex ratio. In order to evaluate the effect of these ligand-like OTCs, we constructed a reconstituted yeast system (CfRE system) based on GAL4 yeast two-hybrid principle using scallop Chlamys farreri retinoid X receptor (CfRXRa) and retinoid X response element (RXRE) to investigate the ligand-induced transactivation of CfRXRa. Responses of CfRXRa to 9-cis retinoic acid (9cRA) and tested four OTCs showed concentration-dependent response which is comparable with reported RXRa in vitro assay of human and gastropods. The detective limits of the CfRE system were found to be 100 nM for 9cRA and 10-1000 nM for the tested OTCs. While the tested non-Sn endocrine disrupting chemicals, including Benzo[a]pyrene, 2,4-Dichlorophenol, Nonylphenol, and Tetrabromobisphenol A, showed no effect on CfRXRa response. The present assay system may provide a valuable tool for screening assessments of unidentified environmental ligand chemicals on bivalve mollusks. It is also useful for comparison of sensitivity differences among species exposed to EDCs.
Collapse
Affiliation(s)
- Anran Zhao
- Ministry of Education, The Key Laboratory of Mariculture (Ocean University of China), Qingdao, 266003, People's Republic of China
| | - Jingjing Miao
- Ministry of Education, The Key Laboratory of Mariculture (Ocean University of China), Qingdao, 266003, People's Republic of China.
| | - Liru Liu
- Ministry of Education, The Key Laboratory of Mariculture (Ocean University of China), Qingdao, 266003, People's Republic of China
| | - Luqing Pan
- Ministry of Education, The Key Laboratory of Mariculture (Ocean University of China), Qingdao, 266003, People's Republic of China
| |
Collapse
|