1
|
Hackney JF, Broatch JE, Dallal RA, Brotherson C, Livingston S, Sabir Z, Reshi SM, Faltermeier Petras SR, Mallick S, Applegate MT, Mellor NJ, Buss K, Blain JM, Wagner CE, Jurutka PW, Marshall PA. Rexinoids Induce Differential Gene Expression in Human Glioblastoma Cells and Protein-Protein Interactions in a Yeast Two-Hybrid System. ACS Chem Neurosci 2024; 15:2897-2915. [PMID: 39012782 DOI: 10.1021/acschemneuro.4c00286] [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: 07/18/2024] Open
Abstract
Rexinoids are compounds that bind to the rexinoid X receptor (RXR) to modulate gene expression and have been proposed as a new class of therapeutics to treat Alzheimer's disease. Different rexinoids will initiate downstream effects that can be quite marked even though such compounds can be structurally similar and have comparable RXR binding affinities. RXR can both homo- and heterodimerize, and these protein-protein interactions and subsequent transactivating potential lead to differential gene expression, depending on the RXR dimeric partner, additional cofactors recruited, and downstream transcription factors that are up- or downregulated. Expression analysis was performed in the U87 human glioblastoma cell line treated with a panel of rexinoids, and our analysis demonstrated that rexinoids with similar RXR EC50 values can have pronounced differences in differential gene expression. Rexinoid binding likely leads to distinctive RXR conformations that cause major downstream gene expression alterations via modulation of RXR interacting proteins. Yeast two-hybrid analysis of RXR bait with two RXR interacting partners demonstrates that rexinoids drive differential binding of RXR to distinctive protein partners. Physiochemical analysis of the rexinoids reveals that the molecules cluster similarly to their gene expression patterns. Thus, rexinoids with similar RXR binding affinities drive differential gene expression by stimulating additional binding patterns in RXR and its homo- and heteropartners, driven by the physicochemical characteristics of these molecules.
Collapse
Affiliation(s)
- Jennifer F Hackney
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Jennifer E Broatch
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Rita A Dallal
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Christian Brotherson
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Sarah Livingston
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Zhela Sabir
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Sabeeha Mushtaq Reshi
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Samantha R Faltermeier Petras
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Sanchita Mallick
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Michael T Applegate
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Nicholas J Mellor
- Genomics Core, Biosciences, Arizona State University, Tempe, Arizona 85281, United States
| | - Kristina Buss
- Genomics Core, Biosciences, Arizona State University, Tempe, Arizona 85281, United States
| | - Joy M Blain
- Genomics Core, Biosciences, Arizona State University, Tempe, Arizona 85281, United States
| | - Carl E Wagner
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Peter W Jurutka
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| | - Pamela A Marshall
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, Arizona 85306, United States
| |
Collapse
|
2
|
Replogle MR, Thompson S, Reis LM, Semina EV. A De Novo Noncoding RARB Variant Associated with Complex Microphthalmia Alters a Putative Regulatory Element. Hum Mutat 2024; 2024:6619280. [PMID: 39450403 PMCID: PMC11501074 DOI: 10.1155/2024/6619280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Retinoic acid receptor beta (RARB) is a transcriptional regulator crucial for coordinating retinoic acid- (RA-) mediated morphogenic movements, cell growth, and differentiation during eye development. Loss- or gain-of-function RARB coding variants have been associated with microphthalmia, coloboma, and anterior segment defects. We identified a de novo variant c.157+1895G>A located within a conserved region (CR1) in the first intron of RARB in an individual with complex microphthalmia and significant global developmental delay. Based on the phenotypic overlap, we further investigated the possible effects of the variant on mRNA splicing and/or transcriptional regulation through in silico and functional studies. In silico analysis identified the possibility of alternative splicing, suggested by one out of three (HSF, SpliceAI, and MaxEntScan) splicing prediction programs, and a strong indication of regulatory function based on publicly available DNase hypersensitivity, histone modification, chromatin folding, and ChIP-seq data sets. Consistent with the predictions of SpliceAI and MaxEntScan, in vitro minigene assays showed no effect on RARB mRNA splicing. Evaluation of CR1 for a regulatory role using luciferase reporter assays in human lens epithelial cells demonstrated a significant increase in the activity of the RARB promoter in the presence of wild-type CR1. This activity was further significantly increased in the presence of CR1 carrying the c.157+1895G>A variant, suggesting that the variant may promote RARB overexpression in human cells. Induction of RARB overexpression in human lens epithelial cells resulted in increased cell proliferation and elevated expression of FOXC1, a known downstream target of RA signaling and a transcription factor whose down- and upregulation is associated with ocular phenotypes overlapping the RARB spectrum. These results support a regulatory role for the CR1 element and suggest that the de novo c.157+1895G>A variant affecting this region may alter the proper regulation of RARB and, as a result, its downstream genes, possibly leading to abnormal development.
Collapse
Affiliation(s)
- Maria R. Replogle
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samuel Thompson
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Linda M. Reis
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Elena V. Semina
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Hospital of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
3
|
Bohn T, Böhm V, Dulińska-Litewka J, Landrier JF, Bánáti D, Kucuk O, Borel P, Canas JA, Rühl R. Is vitamin A an antioxidant? INT J VITAM NUTR RES 2023; 93:481-482. [PMID: 35291873 DOI: 10.1024/0300-9831/a000752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Torsten Bohn
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Volker Böhm
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Germany
| | | | | | - Diána Bánáti
- Faculty of Engineering, University of Szeged, Hungary
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute of Emory University, Druid Hills, Georgia, USA
| | - Patrick Borel
- C2VN, INRAE, INSERM, Aix-Marseille University, Marseille, France
| | - Jose A Canas
- Division of Pediatric Endocrinology, John's Hopkins All Children's Hospital, Saint Petersburg, Florida, USA
| | - Ralph Rühl
- Paprika Bioanalytics BT, Debrecen, Hungary
| |
Collapse
|
4
|
Lavudi K, Nuguri SM, Olverson Z, Dhanabalan AK, Patnaik S, Kokkanti RR. Targeting the retinoic acid signaling pathway as a modern precision therapy against cancers. Front Cell Dev Biol 2023; 11:1254612. [PMID: 37645246 PMCID: PMC10461636 DOI: 10.3389/fcell.2023.1254612] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/28/2023] [Indexed: 08/31/2023] Open
Abstract
Retinoic acid (RA) is a vital metabolite derived from vitamin A. RA plays a prominent role during development, which helps in embryological advancement and cellular differentiation. Mechanistically, RA binds to its definite nuclear receptors including the retinoic acid receptor and retinoid X receptor, thus triggering gene transcription and further consequences in gene regulation. This functional heterodimer activation later results in gene activation/inactivation. Several reports have been published related to the detailed embryonic and developmental role of retinoic acids and as an anti-cancer drug for specific cancers, including acute promyelocytic leukemia, breast cancer, and prostate cancer. Nonetheless, the other side of all-trans retinoic acid (ATRA) has not been explored widely yet. In this review, we focused on the role of the RA pathway and its downstream gene activation in relation to cancer progression. Furthermore, we explored the ways of targeting the retinoic acid pathway by focusing on the dual role of aldehyde dehydrogenase (ALDH) family enzymes. Combination strategies by combining RA targets with ALDH-specific targets make the tumor cells sensitive to the treatment and improve the progression-free survival of the patients. In addition to the genomic effects of ATRA, we also highlighted the role of ATRA in non-canonical mechanisms as an immune checkpoint inhibitor, thus targeting the immune oncological perspective of cancer treatments in the current era. The role of ATRA in activating independent mechanisms is also explained in this review. This review also highlights the current clinical trials of ATRA in combination with other chemotherapeutic drugs and explains the future directional insights related to ATRA usage.
Collapse
Affiliation(s)
- Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Shreya Madhav Nuguri
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Zianne Olverson
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Anantha Krishna Dhanabalan
- Centre for Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, India
| | - Srinivas Patnaik
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Rekha Rani Kokkanti
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam, Tirupati, Andhra Pradesh, India
| |
Collapse
|
5
|
Choi WJ, Haratipour Z, Blind RD. Full-length nuclear receptor allosteric regulation. J Lipid Res 2023; 64:100406. [PMID: 37356665 PMCID: PMC10388211 DOI: 10.1016/j.jlr.2023.100406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/27/2023] Open
Abstract
Nuclear receptors are a superfamily of transcription factors regulated by a wide range of lipids that include phospholipids, fatty acids, heme-based metabolites, and cholesterol-based steroids. Encoded as classic two-domain modular transcription factors, nuclear receptors possess a DNA-binding domain (DBD) and a lipid ligand-binding domain (LBD) containing a transcriptional activation function. Decades of structural studies on the isolated LBDs of nuclear receptors established that lipid-ligand binding allosterically regulates the conformation of the LBD, regulating transcriptional coregulator recruitment and thus nuclear receptor function. These structural studies have aided the development of several FDA-approved drugs, highlighting the importance of understanding the structure-function relationships between lipids and nuclear receptors. However, there are few published descriptions of full-length nuclear receptor structure and even fewer descriptions of how lipids might allosterically regulate full-length structure. Here, we examine multidomain interactions based on the published full-length nuclear receptor structures, evaluating the potential of interdomain interfaces within these nuclear receptors to act as inducible sites of allosteric regulation by lipids.
Collapse
Affiliation(s)
- Woong Jae Choi
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zeinab Haratipour
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Center for Structural Biology, Nashville, TN, USA; Program in Precision Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Raymond D Blind
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt University Center for Structural Biology, Nashville, TN, USA; Program in Precision Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Diabetes Research and Training Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
6
|
An Isochroman Analog of CD3254 and Allyl-, Isochroman-Analogs of NEt-TMN Prove to Be More Potent Retinoid-X-Receptor (RXR) Selective Agonists Than Bexarotene. Int J Mol Sci 2022; 23:ijms232416213. [PMID: 36555852 PMCID: PMC9782500 DOI: 10.3390/ijms232416213] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Bexarotene is an FDA-approved drug for the treatment of cutaneous T-cell lymphoma (CTCL); however, its use provokes or disrupts other retinoid-X-receptor (RXR)-dependent nuclear receptor pathways and thereby incites side effects including hypothyroidism and raised triglycerides. Two novel bexarotene analogs, as well as three unique CD3254 analogs and thirteen novel NEt-TMN analogs, were synthesized and characterized for their ability to induce RXR agonism in comparison to bexarotene (1). Several analogs in all three groups possessed an isochroman ring substitution for the bexarotene aliphatic group. Analogs were modeled for RXR binding affinity, and EC50 as well as IC50 values were established for all analogs in a KMT2A-MLLT3 leukemia cell line. All analogs were assessed for liver-X-receptor (LXR) activity in an LXRE system to gauge the potential for the compounds to provoke raised triglycerides by increasing LXR activity, as well as to drive LXRE-mediated transcription of brain ApoE expression as a marker for potential therapeutic use in neurodegenerative disorders. Preliminary results suggest these compounds display a broad spectrum of off-target activities. However, many of the novel compounds were observed to be more potent than 1. While some RXR agonists cross-signal the retinoic acid receptor (RAR), many of the rexinoids in this work displayed reduced RAR activity. The isochroman group did not appear to substantially reduce RXR activity on its own. The results of this study reveal that modifying potent, selective rexinoids like bexarotene, CD3254, and NEt-TMN can provide rexinoids with increased RXR selectivity, decreased potential for cross-signaling, and improved anti-proliferative characteristics in leukemia models compared to 1.
Collapse
|
7
|
Jurutka PW, di Martino O, Reshi S, Mallick S, Sabir ZL, Staniszewski LJP, Warda A, Maiorella EL, Minasian A, Davidson J, Ibrahim SJ, Raban S, Haddad D, Khamisi M, Suban SL, Dawson BJ, Candia R, Ziller JW, Lee MY, Liu C, Liu W, Marshall PA, Welch JS, Wagner CE. Modeling, Synthesis, and Biological Evaluation of Potential Retinoid-X-Receptor (RXR) Selective Agonists: Analogs of 4-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahyro-2-naphthyl)ethynyl]benzoic Acid (Bexarotene) and 6-(Ethyl(4-isobutoxy-3-isopropylphenyl)amino)nicotinic Acid (NEt-4IB). Int J Mol Sci 2021; 22:ijms222212371. [PMID: 34830251 PMCID: PMC8624485 DOI: 10.3390/ijms222212371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/08/2021] [Accepted: 11/12/2021] [Indexed: 12/05/2022] Open
Abstract
Five novel analogs of 6-(ethyl)(4-isobutoxy-3-isopropylphenyl)amino)nicotinic acid—or NEt-4IB—in addition to seven novel analogs of 4-[1-(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethynyl]benzoic acid (bexarotene) were prepared and evaluated for selective retinoid-X-receptor (RXR) agonism alongside bexarotene (1), a FDA-approved drug for cutaneous T-cell lymphoma (CTCL). Bexarotene treatment elicits side-effects by provoking or disrupting other RXR-dependent pathways. Analogs were assessed by the modeling of binding to RXR and then evaluated in a human cell-based RXR-RXR mammalian-2-hybrid (M2H) system as well as a RXRE-controlled transcriptional system. The analogs were also tested in KMT2A-MLLT3 leukemia cells and the EC50 and IC50 values were determined for these compounds. Moreover, the analogs were assessed for activation of LXR in an LXRE system as drivers of ApoE expression and subsequent use as potential therapeutics in neurodegenerative disorders, and the results revealed that these compounds exerted a range of differential LXR-RXR activation and selectivity. Furthermore, several of the novel analogs in this study exhibited reduced RARE cross-signaling, implying RXR selectivity. These results demonstrate that modification of partial agonists such as NEt-4IB and potent rexinoids such as bexarotene can lead to compounds with improved RXR selectivity, decreased cross-signaling of other RXR-dependent nuclear receptors, increased LXRE-heterodimer selectivity, and enhanced anti-proliferative potential in leukemia cell lines compared to therapeutics such as 1.
Collapse
Affiliation(s)
- Peter W. Jurutka
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
- Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Orsola di Martino
- Department of Internal Medicine, Washington University, St. Louis, MO 63110, USA; (O.d.M.); (J.S.W.)
| | - Sabeeha Reshi
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Sanchita Mallick
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Zhela L. Sabir
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Lech J. P. Staniszewski
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Ankedo Warda
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
- Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Emma L. Maiorella
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Ani Minasian
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Jesse Davidson
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Samir J. Ibrahim
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - San Raban
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Dena Haddad
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Madleen Khamisi
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Stephanie L. Suban
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Bradley J. Dawson
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Riley Candia
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - Joseph W. Ziller
- Department of Chemistry, University of California, Irvine, CA 92697, USA;
| | - Ming-Yue Lee
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85201, USA; (M.-Y.L.); (C.L.); (W.L.)
| | - Chang Liu
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85201, USA; (M.-Y.L.); (C.L.); (W.L.)
| | - Wei Liu
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85201, USA; (M.-Y.L.); (C.L.); (W.L.)
| | - Pamela A. Marshall
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
| | - John S. Welch
- Department of Internal Medicine, Washington University, St. Louis, MO 63110, USA; (O.d.M.); (J.S.W.)
| | - Carl E. Wagner
- School of Mathematical and Natural Sciences, Arizona State University, Glendale, AZ 85306, USA; (P.W.J.); (S.R.); (S.M.); (Z.L.S.); (L.J.P.S.); (A.W.); (E.L.M.); (A.M.); (J.D.); (S.J.I.); (S.R.); (D.H.); (M.K.); (S.L.S.); (B.J.D.); (R.C.); (P.A.M.)
- Correspondence: ; Tel.: +1-602-543-6937
| |
Collapse
|
8
|
Afshari-Kaveh M, Abbasalipourkabir R, Nourian A, Ziamajidi N. The Protective Effects of Vitamins A and E on Titanium Dioxide Nanoparticles (nTiO2)-Induced Oxidative Stress in the Spleen Tissues of Male Wistar Rats. Biol Trace Elem Res 2021; 199:3677-3687. [PMID: 33210191 DOI: 10.1007/s12011-020-02487-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
Titanium dioxide nanoparticles (nTiO2) can accumulate in different tissues and damage them with oxidative stress induction. Different components with antioxidant capacity can protect the tissues. So in this study, the protective effects of vitamin A and E on the nTiO2-induced oxidative stress in rats' spleen tissues were examined. Thirty-six male Wistar rats were randomly divided into 6 groups: Control 1 (received water), nTiO2, nTiO2 + vitamin E, nTiO2 + vitamin A, nTiO2 + vitamin A and E, and Control 2 (received olive oil). To investigate the status of oxidative stress, total antioxidant capacity (TAC), total oxidant status (TOS), and lipid peroxidation (LPO) were determined in spleen tissue as well as the activities of antioxidant enzymes, including glutathione peroxidase (GPx) and superoxide dismutase (SOD). Also, the gene expression of GPx, SOD, and nuclear factor-E2-related factor-2 (Nrf-2) were determined by qRT-PCR. To evaluate the spleen histopathological changes, H&E staining was carried out. nTiO2 significantly increased TOS and LPO levels, whereas it decreased TAC level, GPx and SOD activities, and gene expression of GPx, SOD, and Nrf-2 in spleen tissues of rats compared with controls (p < 0.05). In vitamin-treated rats, the levels of TOS and LPO significantly decreased, and the level of TAC, the activities of GPx and SOD, and the gene expression of GPx, SOD, and Nrf-2 increased compared to nTiO2 group (p < 0.05). These parameters are maintained near to normal levels. Histological findings confirmed the protective effects of these vitamins on tissue damage caused by nTiO2. Vitamin A and E can protect the spleen tissues from nTiO2-induced oxidative stress.
Collapse
Affiliation(s)
- Mozhgan Afshari-Kaveh
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Nourian
- Department of Pathobiology, Faculty of Paraveterinary Medicine, Bu-Ali Sina University, Hamedan, Iran
| | - Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
9
|
Kim YJ, Park S, Ha T, Kim S, Lim S, You H, Kim JW. Retinoid Metabolism in the Degeneration of Pten-Deficient Mouse Retinal Pigment Epithelium. Mol Cells 2021; 44:613-622. [PMID: 34376625 PMCID: PMC8424139 DOI: 10.14348/molcells.2021.0138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 11/27/2022] Open
Abstract
In vertebrate eyes, the retinal pigment epithelium (RPE) provides structural and functional homeostasis to the retina. The RPE takes up retinol (ROL) to be dehydrogenated and isomerized to 11-cis-retinaldehyde (11-cis-RAL), which is a functional photopigment in mammalian photoreceptors. As excessive ROL is toxic, the RPE must also establish mechanisms to protect against ROL toxicity. Here, we found that the levels of retinol dehydrogenases (RDHs) are commonly decreased in phosphatase tensin homolog (Pten)-deficient mouse RPE, which degenerates due to elevated ROL and that can be rescued by feeding a ROL-free diet. We also identified that RDH gene expression is regulated by forkhead box O (FOXO) transcription factors, which are inactivated by hyperactive Akt in the Pten-deficient mouse RPE. Together, our findings suggest that a homeostatic pathway comprising PTEN, FOXO, and RDH can protect the RPE from ROL toxicity.
Collapse
Affiliation(s)
- You-Joung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Sooyeon Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Taejeong Ha
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Seungbeom Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Soyeon Lim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Han You
- School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
10
|
Abdelwahab EMM, Bovari-Biri J, Smuk G, Harko T, Fillinger J, Moldvay J, Krymskaya VP, Pongracz JE. Normalization of Enzyme Expression and Activity Regulating Vitamin A Metabolism Increases RAR-Beta Expression and Reduces Cellular Migration and Proliferation in Diseases Caused by Tuberous Sclerosis Gene Mutations. Front Oncol 2021; 11:644592. [PMID: 34178631 PMCID: PMC8226169 DOI: 10.3389/fonc.2021.644592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/25/2021] [Indexed: 11/15/2022] Open
Abstract
Background Mutation in a tuberous sclerosis gene (TSC1 or 2) leads to continuous activation of the mammalian target of rapamycin (mTOR). mTOR activation alters cellular including vitamin A metabolism and retinoic acid receptor beta (RARβ) expression. The goal of the present study was to investigate the molecular connection between vitamin A metabolism and TSC mutation. We also aimed to investigate the effect of the FDA approved drug rapamycin and the vitamin A metabolite retinoic acid (RA) in cell lines with TSC mutation. Methods Expression and activity of vitamin A associated metabolic enzymes and RARβ were assessed in human kidney angiomyolipoma derived cell lines, primary lymphangioleiomyomatosis (LAM) tissue derived LAM cell lines. RARβ protein levels were also tested in primary LAM lung tissue sections. TaqMan arrays, enzyme activities, qRT-PCRs, immunohistochemistry, immunofluorescent staining, and western blotting were performed and analysed. The functional effects of retinoic acid (RA) and rapamycin were tested in a scratch and a BrDU assay to assess cell migration and proliferation. Results Metabolic enzyme arrays revealed a general deregulation of many enzymes involved in vitamin A metabolism including aldehyde dehydrogenases (ALDHs), alcohol dehydrogenases (ADHs) and Cytochrome P450 2E1 (CYP2E1). Furthermore, RARβ downregulation was a characteristic feature of all TSC-deficient cell lines and primary tissues. Combination of the two FDA approved drugs -RA for acute myeloid leukaemia and rapamycin for TSC mutation- normalised ALDH and ADH expression and activity, restored RARβ expression and reduced cellular proliferation and migration. Conclusion Deregulation of vitamin A metabolizing enzymes is a feature of TSC mutation. RA can normalize RARβ levels and limit cell migration but does not have a significant effect on proliferation. Based on our data, translational studies could confirm whether combination of RA with reduced dosage of rapamycin would have more beneficial effects to higher dosage of rapamycin monotherapy meanwhile reducing adverse effects of rapamycin for patients with TSC mutation.
Collapse
Affiliation(s)
| | - Judit Bovari-Biri
- Departments of Pharmaceutical Biotechnology, University of Pecs, Pecs, Hungary.,Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Gabor Smuk
- Department of Pathology, University of Pecs, Pecs, Hungary
| | - Tunde Harko
- Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Janos Fillinger
- Department of Pathology, Semmelweis University, Budapest, Hungary.,Department of Pulmonology, National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Judit Moldvay
- Department of Pathology, Semmelweis University, Budapest, Hungary.,Department of Pulmonology, National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Vera P Krymskaya
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Judit E Pongracz
- Departments of Pharmaceutical Biotechnology, University of Pecs, Pecs, Hungary.,Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| |
Collapse
|
11
|
Song Y, Kurose A, Li R, Takeda T, Onomura Y, Koga T, Mutoh J, Ishida T, Tanaka Y, Ishii Y. Ablation of Selenbp1 Alters Lipid Metabolism via the Pparα Pathway in Mouse Kidney. Int J Mol Sci 2021; 22:ijms22105334. [PMID: 34069420 PMCID: PMC8159118 DOI: 10.3390/ijms22105334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/10/2021] [Accepted: 05/15/2021] [Indexed: 12/12/2022] Open
Abstract
Selenium-binding protein 1 (Selenbp1) is a 2,3,7,8-tetrechlorodibenzo-p-dioxin inducible protein whose function is yet to be comprehensively elucidated. As the highly homologous isoform, Selenbp2, is expressed at low levels in the kidney, it is worthwhile comparing wild-type C57BL mice and Selenbp1-deficient mice under dioxin-free conditions. Accordingly, we conducted a mouse metabolomics analysis under non-dioxin-treated conditions. DNA microarray analysis was performed based on observed changes in lipid metabolism-related factors. The results showed fluctuations in the expression of numerous genes. Real-time RT-PCR confirmed the decreased expression levels of the cytochrome P450 4a (Cyp4a) subfamily, known to be involved in fatty acid ω- and ω-1 hydroxylation. Furthermore, peroxisome proliferator-activated receptor-α (Pparα) and retinoid-X-receptor-α (Rxrα), which form a heterodimer with Pparα to promote gene expression, were simultaneously reduced. This indicated that reduced Cyp4a expression was mediated via decreased Pparα and Rxrα. In line with this finding, increased levels of leukotrienes and prostaglandins were detected. Conversely, decreased hydrogen peroxide levels and reduced superoxide dismutase (SOD) activity supported the suppression of the renal expression of Sod1 and Sod2 in Selenbp1-deficient mice. Therefore, we infer that ablation of Selenbp1 elicits oxidative stress caused by increased levels of superoxide anions, which alters lipid metabolism via the Pparα pathway.
Collapse
Affiliation(s)
- Yingxia Song
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (Y.S.); (A.K.); (R.L.); (T.T.); (Y.O.)
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Atsushi Kurose
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (Y.S.); (A.K.); (R.L.); (T.T.); (Y.O.)
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Renshi Li
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (Y.S.); (A.K.); (R.L.); (T.T.); (Y.O.)
| | - Tomoki Takeda
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (Y.S.); (A.K.); (R.L.); (T.T.); (Y.O.)
| | - Yuko Onomura
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (Y.S.); (A.K.); (R.L.); (T.T.); (Y.O.)
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Takayuki Koga
- Laboratory of Hygienic Chemistry, Daiichi University of Pharmacy, 22-1 Tamagawa-cho, Minami-ku, Fukuoka 815-8511, Japan;
| | - Junpei Mutoh
- Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Daigakudori 1-1-1, Sanyo-Onoda 756-0884, Japan;
| | - Takumi Ishida
- School of Pharmacy, International University of Health and Welfare Fukuoka, Ohkawa, Fukuoka 831-8501, Japan;
| | - Yoshitaka Tanaka
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Yuji Ishii
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (Y.S.); (A.K.); (R.L.); (T.T.); (Y.O.)
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
- Correspondence: ; Tel.: +81-92-642-6586
| |
Collapse
|
12
|
Multi-species transcriptome meta-analysis of the response to retinoic acid in vertebrates and comparative analysis of the effects of retinol and retinoic acid on gene expression in LMH cells. BMC Genomics 2021; 22:146. [PMID: 33653267 PMCID: PMC7923837 DOI: 10.1186/s12864-021-07451-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/18/2021] [Indexed: 12/21/2022] Open
Abstract
Background Retinol (RO) and its active metabolite retinoic acid (RA) are major regulators of gene expression in vertebrates and influence various processes like organ development, cell differentiation, and immune response. To characterize a general transcriptomic response to RA-exposure in vertebrates, independent of species- and tissue-specific effects, four publicly available RNA-Seq datasets from Homo sapiens, Mus musculus, and Xenopus laevis were analyzed. To increase species and cell-type diversity we generated RNA-seq data with chicken hepatocellular carcinoma (LMH) cells. Additionally, we compared the response of LMH cells to RA and RO at different time points. Results By conducting a transcriptome meta-analysis, we identified three retinoic acid response core clusters (RARCCs) consisting of 27 interacting proteins, seven of which have not been associated with retinoids yet. Comparison of the transcriptional response of LMH cells to RO and RA exposure at different time points led to the identification of non-coding RNAs (ncRNAs) that are only differentially expressed (DE) during the early response. Conclusions We propose that these RARCCs stand on top of a common regulatory RA hierarchy among vertebrates. Based on the protein sets included in these clusters we were able to identify an RA-response cluster, a control center type cluster, and a cluster that directs cell proliferation. Concerning the comparison of the cellular response to RA and RO we conclude that ncRNAs play an underestimated role in retinoid-mediated gene regulation. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07451-2.
Collapse
|
13
|
Szymański Ł, Skopek R, Palusińska M, Schenk T, Stengel S, Lewicki S, Kraj L, Kamiński P, Zelent A. Retinoic Acid and Its Derivatives in Skin. Cells 2020; 9:E2660. [PMID: 33322246 PMCID: PMC7764495 DOI: 10.3390/cells9122660] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/29/2020] [Accepted: 12/07/2020] [Indexed: 12/30/2022] Open
Abstract
The retinoids are a group of compounds including vitamin A and its active metabolite all-trans-retinoic acid (ATRA). Retinoids regulate a variety of physiological functions in multiple organ systems, are essential for normal immune competence, and are involved in the regulation of cell growth and differentiation. Vitamin A derivatives have held promise in cancer treatment and ATRA is used in differentiation therapy of acute promyelocytic leukemia (APL). ATRA and other retinoids have also been successfully applied in a variety of dermatological conditions such as skin cancer, psoriasis, acne, and ichthyosis. Moreover, modulation of retinoic acid receptors and retinoid X (or rexinoid) receptors function may affect dermal cells. The studies using complex genetic models with various combinations of retinoic acid receptors (RARs) and retinoid X (or rexinoid) receptors (RXRs) indicate that retinoic acid and its derivatives have therapeutic potential for a variety of serious dermatological disorders including some malignant conditions. Here, we provide a synopsis of the main advances in understanding the role of ATRA and its receptors in dermatology.
Collapse
Affiliation(s)
- Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (Ł.S.); (R.S.); (M.P.)
| | - Rafał Skopek
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (Ł.S.); (R.S.); (M.P.)
| | - Małgorzata Palusińska
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (Ł.S.); (R.S.); (M.P.)
| | - Tino Schenk
- Department of Hematology/Oncology, Clinic of Internal Medicine II, Jena University Hospital, 07747 Jena, Germany;
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine Jena (CMB), Jena University Hospital, 07747 Jena, Germany
| | - Sven Stengel
- Department of Internal Medicine IV, Division of Gastroenterology, Hepatology and Infectious Disease, Jena University Hospital, Friedrich Schiller University of Jena, 07747 Jena, Germany;
| | - Sławomir Lewicki
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology, 01-163 Warsaw, Poland
- Department of Medicine, Faculty of Medical Sciences and Health Sciences, Kazimierz Pulaski University of Technology and Humanities, 26-600 Radom, Poland
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, 01-163 Warsaw, Poland;
| | - Paweł Kamiński
- Department of Gynecology and Oncological Gynecology, Military Institute of Medicine, 01-163 Warsaw, Poland;
| | - Arthur Zelent
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (Ł.S.); (R.S.); (M.P.)
| |
Collapse
|
14
|
Reynolds K, Zhang S, Sun B, Garland M, Ji Y, Zhou CJ. Genetics and signaling mechanisms of orofacial clefts. Birth Defects Res 2020; 112:1588-1634. [PMID: 32666711 PMCID: PMC7883771 DOI: 10.1002/bdr2.1754] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022]
Abstract
Craniofacial development involves several complex tissue movements including several fusion processes to form the frontonasal and maxillary structures, including the upper lip and palate. Each of these movements are controlled by many different factors that are tightly regulated by several integral morphogenetic signaling pathways. Subject to both genetic and environmental influences, interruption at nearly any stage can disrupt lip, nasal, or palate fusion and result in a cleft. Here, we discuss many of the genetic risk factors that may contribute to the presentation of orofacial clefts in patients, and several of the key signaling pathways and underlying cellular mechanisms that control lip and palate formation, as identified primarily through investigating equivalent processes in animal models, are examined.
Collapse
Affiliation(s)
- Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| | - Shuwen Zhang
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Michael Garland
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| | - Chengji J. Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| |
Collapse
|
15
|
Wang T, Mei J, Li X, Xu X, Ma B, Li W. A novel tsRNA-16902 regulating the adipogenic differentiation of human bone marrow mesenchymal stem cells. Stem Cell Res Ther 2020; 11:365. [PMID: 32831139 PMCID: PMC7444066 DOI: 10.1186/s13287-020-01882-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/18/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Transfer RNA-derived small RNAs (tsRNAs) are a recently discovered form of non-coding RNA capable of regulating myriad physiological processes. The role of tsRNAs in hMSC adipogenic differentiation, however, remains incompletely understood. The purpose of this study was to identify the novel tsRNA-16902 as a regulator of hMSC adipogenic differentiation. METHODS In this study, we conducted transcriptomic sequencing of hMSCs after inducing their adipogenic differentiation, and we were thereby able to clarify the molecular mechanism underlying the role of tsRNA-16902 in this context via a series of molecular biology methods. RESULTS When we knocked down tsRNA-16902 expression, this impaired hMSC adipogenic differentiation and associated marker gene expression. Bioinformatics analyses further revealed tsRNA-16902 to target retinoic acid receptor γ (RARγ). Luciferase reporter assays also confirmed the ability of tsRNA-16902 to bind to the RARγ 3'-untranslated region. Consistent with this, RARγ overexpression led to impaired hMSC adipogenesis. Further analyses revealed that Smad2/3 phosphorylation was increased in cells that either overexpressed RARγ or in which tsRNA-16902 had been knocked down. We also assessed the adipogenic differentiation of hMSCs in which tsRNA-16902 was knocked down and at the same time a Smad2/3 inhibitor was added to disrupt Smad2/3 phosphorylation. The adipogenic differentiation of hMSCs in which tsRNA-16902 was knocked down was further enhanced upon the addition of a Smad2/3 signaling inhibitor relative to tsRNA-16902 knockdown alone. CONCLUSIONS Through a comprehensive profiling analysis of tsRNAs that were differentially expressed in the context of hMSC adipogenic differentiation, we were able to identify tsRNA-16902 as a previously uncharacterized regulator of adipogenesis. tsRNA-16902 is able to regulate hMSC adipogenic differentiation by targeting RARγ via the Smad2/3 signaling pathway. Together, our results may thus highlight novel strategies of value for treating obesity.
Collapse
Affiliation(s)
- Tao Wang
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China.
| | - Jun Mei
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Xingnuan Li
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Xiaoyuan Xu
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China
| | - Baicheng Ma
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China.
| | - Weidong Li
- Key Laboratory of System Bio-medicine of Jiangxi Province, Jiujiang University, Jiujiang, 332000, China.
| |
Collapse
|
16
|
Retinoic acid receptor α as a novel contributor to adrenal cortex structure and function through interactions with Wnt and Vegfa signalling. Sci Rep 2019; 9:14677. [PMID: 31605007 PMCID: PMC6789122 DOI: 10.1038/s41598-019-50988-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/17/2019] [Indexed: 01/09/2023] Open
Abstract
Primary aldosteronism (PA) is the most frequent form of secondary arterial hypertension. Mutations in different genes increase aldosterone production in PA, but additional mechanisms may contribute to increased cell proliferation and aldosterone producing adenoma (APA) development. We performed transcriptome analysis in APA and identified retinoic acid receptor alpha (RARα) signaling as a central molecular network involved in nodule formation. To understand how RARα modulates adrenal structure and function, we explored the adrenal phenotype of male and female Rarα knockout mice. Inactivation of Rarα in mice led to significant structural disorganization of the adrenal cortex in both sexes, with increased adrenal cortex size in female mice and increased cell proliferation in males. Abnormalities of vessel architecture and extracellular matrix were due to decreased Vegfa expression and modifications in extracellular matrix components. On the molecular level, Rarα inactivation leads to inhibition of non-canonical Wnt signaling, without affecting the canonical Wnt pathway nor PKA signaling. Our study suggests that Rarα contributes to the maintenance of normal adrenal cortex structure and cell proliferation, by modulating Wnt signaling. Dysregulation of this interaction may contribute to abnormal cell proliferation, creating a propitious environment for the emergence of specific driver mutations in PA.
Collapse
|
17
|
Effects of vitamin A and vitamin E on attenuation of titanium dioxide nanoparticles-induced toxicity in the liver of male Wistar rats. Mol Biol Rep 2019; 46:2919-2932. [DOI: 10.1007/s11033-019-04752-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 03/08/2019] [Indexed: 12/12/2022]
|
18
|
Sugrue KF, Sarkar AA, Leatherbury L, Zohn IE. The ubiquitin ligase HECTD1 promotes retinoic acid signaling required for development of the aortic arch. Dis Model Mech 2019; 12:dmm.036491. [PMID: 30578278 PMCID: PMC6361158 DOI: 10.1242/dmm.036491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022] Open
Abstract
The development of the aortic arch is a complex process that involves remodeling of the bilaterally symmetrical pharyngeal arch arteries (PAAs) into the mature asymmetric aortic arch. Retinoic acid signaling is a key regulator of this process by directing patterning of the second heart field (SHF), formation of the caudal PAAs and subsequent remodeling of the PAAs to form the aortic arch. Here, we identify the HECTD1 ubiquitin ligase as a novel modulator of retinoic acid signaling during this process. Hectd1opm/opm homozygous mutant embryos show a spectrum of aortic arch abnormalities that occur following loss of 4th PAAs and increased SHF marker expression. This sequence of defects is similar to phenotypes observed in mutant mouse models with reduced retinoic acid signaling. Importantly, HECTD1 binds to and influences ubiquitination of the retinoic acid receptor, alpha (RARA). Furthermore, reduced activation of a retinoic acid response element (RARE) reporter is detected in Hectd1 mutant cells and embryos. Interestingly, Hectd1opm/+ heterozygous embryos exhibit reduced retinoic acid signaling, along with intermediate increased expression of SHF markers; however, heterozygotes show normal development of the aortic arch. Decreasing retinoic acid synthesis by reducing Raldh2 (also known as Aldh1a2) gene dosage in Hectd1opm/+ heterozygous embryos reveals a genetic interaction. Double heterozygous embryos show hypoplasia of the 4th PAA and increased incidence of a benign aortic arch variant, in which the transverse arch between the brachiocephalic and left common carotid arteries is shortened. Together, our data establish that HECTD1 is a novel regulator of retinoic acid signaling required for proper aortic arch development. Editor's choice: The HECTD1 ubiquitin ligase is a novel modulator of retinoic acid signaling during aortic arch development and provides a model for complex interactions underlying variations in aortic arch development.
Collapse
Affiliation(s)
- Kelsey F Sugrue
- Institute for Biomedical Sciences, The George Washington University, Washington, DC 20037, USA.,Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Anjali A Sarkar
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Linda Leatherbury
- Children's National Heart Institute, Children's National Health System, George Washington University School of Medicine, Washington, DC 20010, USA
| | - Irene E Zohn
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| |
Collapse
|
19
|
Wagner CE, Jurutka PW. Methods to Generate an Array of Novel Rexinoids by SAR on a Potent Retinoid X Receptor Agonist: A Case Study with NEt-TMN. Methods Mol Biol 2019; 2019:109-121. [PMID: 31359392 DOI: 10.1007/978-1-4939-9585-1_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The methods described in this chapter concern procedures for the design, synthesis, and in vitro biological evaluation of an array of potent retinoid-X-receptor (RXR) agonists employing 6-(ethyl(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)amino)nicotinic acid (NEt-TMN), and recently reported NEt-TMN analogs, as a case study. These methods have been extensively applied beyond the present case study to generate several analogs of other potent RXR agonists (rexinoids), particularly the RXR agonist known as bexarotene (Bex), a Food and Drug Administration (FDA) approved drug for cutaneous T-cell lymphoma that is also often prescribed, off-label, for breast, lung, and other human cancers. Common side effects with Bex treatment include hypertriglyceridemia and hypothyroidism, because of off-target activation or inhibition of other nuclear receptor pathways impacted by RXR. Because rexinoids are often selective for RXR, versus the retinoic-acid-receptor (RAR), cutaneous toxicity is often avoided as a side effect for rexinoid treatment. Several other potent RXR agonists, and their analogs, have been reported in the literature and rigorously evaluated (often in comparison to Bex) as potential cancer therapeutics with unique activity and side-effect profiles. Some of the more prominent examples include LGD100268, CD3254, and 9-cis-UAB30, to name only a few. Hence, the methods described herein are more widely applicable to a diverse array of RXR agonists.In terms of design, the structure-activity relationship (SAR) study is usually performed by modifying three distinct areas of the rexinoid base structure, either of the nonpolar or polar sides of the rexinoid and/or the linkage that joins them. For the synthesis of the modified base-structure analogs, often identical synthetic strategies used to access the base-structure are applied; however, reasonable alternative synthetic routes may need to be explored if the modified analog intermediates encounter bottlenecks where yields are negligible for a given step in the base-structure route. In fact, this particular problem was encountered and successfully resolved in our case study for generating an array of NEt-TMN analogs.
Collapse
Affiliation(s)
- Carl E Wagner
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, USA.
| | - Peter W Jurutka
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, USA
| |
Collapse
|
20
|
Ciccone V, Terzuoli E, Donnini S, Giachetti A, Morbidelli L, Ziche M. Stemness marker ALDH1A1 promotes tumor angiogenesis via retinoic acid/HIF-1α/VEGF signalling in MCF-7 breast cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:311. [PMID: 30541574 PMCID: PMC6291966 DOI: 10.1186/s13046-018-0975-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022]
Abstract
Background Aldehyde dehydrogenase 1A1 (ALDH1A1), a member of aldehyde dehydrogenase family, is a marker of stemness in breast cancer. During tumor progression cancer stem cells (CSCs) have been reported to secrete angiogenic factors to orchestrate the formation of pathological angiogenesis. This vasculature can represent the source of self-renewal of CSCs and the route for further tumor spreading. The aim of the present study has been to assess whether ALDH1A1 controls the output of angiogenic factors in breast cancer cells and regulates tumor angiogenesis in a panel of in vitro and in vivo models. Methods Stemness status of breast cancer cells was evaluated by the ability to form turmorspheres in vitro. A transwell system was used to assess the angiogenic features of human umbilical vein endothelial cells (HUVEC) when co-cultured with breast cancer cells MCF-7 harboring different levels of ALDH1A1. Under these conditions, we survey endothelial proliferation, migration, tube formation and permeability. Moreover, in vivo, MCF-7 xenografts in immunodeficient mice allow to evaluate blood flow, expression of angiogenic factors and microvascular density (MVD). Results In MCF-7 we observed that ALDH1A1 activity conferred stemness property and its expression correlated with an activation of angiogenic factors. In particular we observed a significant upregulation of hypoxia inducible factor-1α (HIF-1α) and proangiogenic factors, such as vascular endothelial growth factor (VEGF). High levels of ALDH1A1, through the retinoic acid pathway, were significantly associated with VEGF-mediated angiogenesis in vitro. Co-culture of HUVEC with ALDH1A1 expressing tumor cells promoted endothelial proliferation, migration, tube formation and permeability. Conversely, downregulation of ALDH1A1 in MCF-7 resulted in reduction of proangiogenic factor release/expression and impaired HUVEC angiogenic functions. In vivo, when subcutaneously implanted in immunodeficient mice, ALDH1A1 overexpressing breast tumor cells displayed a higher expression of VEGF and MVD. Conclusion In breast tumors, ALDH1A1 expression primes a permissive microenvironment by promoting tumor angiogenesis via retinoic acid dependent mechanism. In conclusion, ALDH1A1 might be associated to progression and diffusion of breast cancer. Electronic supplementary material The online version of this article (10.1186/s13046-018-0975-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valerio Ciccone
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Erika Terzuoli
- Department of Medicine, Surgery and Neuroscience, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Antonio Giachetti
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100, Siena, Italy.
| | - Marina Ziche
- Department of Medicine, Surgery and Neuroscience, University of Siena, Via A. Moro 2, 53100, Siena, Italy.
| |
Collapse
|
21
|
Hanish BJ, Hackney Price JF, Kaneko I, Ma N, van der Vaart A, Wagner CE, Jurutka PW, Marshall PA. A novel gene expression analytics-based approach to structure aided design of rexinoids for development as next-generation cancer therapeutics. Steroids 2018; 135:36-49. [PMID: 29704526 PMCID: PMC5977990 DOI: 10.1016/j.steroids.2018.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/09/2018] [Accepted: 04/18/2018] [Indexed: 12/20/2022]
Abstract
Rexinoids are powerful ligands that bind to retinoid-X-receptors (RXRs) and show great promise as therapeutics for a wide range of diseases, including cancer. However, only one rexinoid, bexarotene (Targretin TM) has been successfully transitioned from the bench to the clinic and used to treat cutaneous T-cell lymphoma (CTCL). Our goal is to develop novel potent rexinoids with a less untoward side effect profile than bexarotene. To this end, we have synthesized a wide array of rexinoids with EC50 values and biological activity similar to bexarotene. In order to determine their suitability for additional downstream analysis, and to identify potential candidate analogs for clinical translation, we treated human CTCL cells in culture and employed microarray technology to assess gene expression profiles. We analyzed twelve rexinoids and found they could be stratified into three distinct categories based on their gene expression: similar to bexarotene, moderately different from bexarotene, and substantially different from bexarotene. Surprisingly, small changes in the structure of the bexarotene parent compound led to marked differences in gene expression profiles. Furthermore, specific analogs diverged markedly from our hypothesis in expression of genes expected to be important for therapeutic promise. However, promoter analysis of genes whose expression was analyzed indicates general regulatory trends along structural frameworks. Our results suggest that certain structural motifs, particularly the basic frameworks found in analog 4 and analog 9, represent important starting points to exploit in generating additional rexinoids for future study and therapeutic applications.
Collapse
Affiliation(s)
- Bentley J Hanish
- New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, United States
| | - Jennifer F Hackney Price
- New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, United States
| | - Ichiro Kaneko
- New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, United States; University of Arizona College of Medicine-Phoenix, Department of Basic Medical Sciences, Phoenix, AZ, United States; Department of Molecular Nutrition, Institution of Health Bioscience, University of Tokushima Graduate School, Kuramoto-cho, Japan
| | - Ning Ma
- Department of Chemistry, University of South Florida, Tampa, FL 33620, United States
| | - Arjan van der Vaart
- Department of Chemistry, University of South Florida, Tampa, FL 33620, United States
| | - Carl E Wagner
- New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, United States
| | - Peter W Jurutka
- New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, United States; University of Arizona College of Medicine-Phoenix, Department of Basic Medical Sciences, Phoenix, AZ, United States; University of Arizona Cancer Center, Tucson, AZ, United States
| | - Pamela A Marshall
- New College of Interdisciplinary Arts and Sciences, Arizona State University, Glendale, AZ, United States.
| |
Collapse
|
22
|
Baba S, Pandith A, Shah Z, Baba R. Pathogenetic implication of fusion genes in acute promyelocytic leukemia and their diagnostic utility. Clin Genet 2018; 95:41-52. [DOI: 10.1111/cge.13372] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 01/16/2023]
Affiliation(s)
- S.M. Baba
- Department of Immunology and Molecular MedicineSher‐I‐Kashmir Institute of Medical Sciences Srinagar India
| | - A.A. Pandith
- Advanced Centre for Human GeneticsSher‐I‐Kashmir Institute of Medical Sciences Srinagar India
| | - Z.A. Shah
- Department of Immunology and Molecular MedicineSher‐I‐Kashmir Institute of Medical Sciences Srinagar India
| | - R.A. Baba
- Department of Immunology and Molecular MedicineSher‐I‐Kashmir Institute of Medical Sciences Srinagar India
| |
Collapse
|
23
|
Nair PR, Alvey C, Jin X, Irianto J, Ivanovska I, Discher DE. Filomicelles Deliver a Chemo-Differentiation Combination of Paclitaxel and Retinoic Acid That Durably Represses Carcinomas in Liver to Prolong Survival. Bioconjug Chem 2018; 29:914-927. [PMID: 29451777 DOI: 10.1021/acs.bioconjchem.7b00816] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Drug resistance and relapse is common in cancer treatments with chemotherapeutics, and while drug combinations with naturally occurring, differentiation-inducing retinoic acid (RA) provide remission-free cures for one type of liquid tumor, solid tumors present major problems for delivery. Here, inspired by filoviruses that can be microns in length, flexible filomicelles that self-assemble from an amphiphilic block copolymer (PEG-PCL) are shown to effectively deliver RA and paclitaxel (TAX) to several solid tumor models, particularly in the liver. These hydrophobic compounds synergistically load into the cores of the elongated micelles, and the coloaded micelles prove most effective at causing cell death, ploidy, and durable regression of tumors compared to free drugs or to separately loaded drugs. RA-TAX filomicelles also reduce mortality of human lung or liver derived cancers engrafted at liver, intraperitoneal, and subcutaneous sites in immunodeficient mice. In vitro studies show that the dual drug micelles effectively suppress proliferation while upregulating a generic differentiation marker. The results highlight the potency of dual-loaded filomicelles in killing cancer cells or else driving their differentiation away from growth.
Collapse
Affiliation(s)
- Praful R Nair
- NanoBioPolymers Lab, and Physical Sciences Oncology Center @ Penn , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Cory Alvey
- NanoBioPolymers Lab, and Physical Sciences Oncology Center @ Penn , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Xiaoling Jin
- NanoBioPolymers Lab, and Physical Sciences Oncology Center @ Penn , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Jerome Irianto
- NanoBioPolymers Lab, and Physical Sciences Oncology Center @ Penn , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Irena Ivanovska
- NanoBioPolymers Lab, and Physical Sciences Oncology Center @ Penn , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Dennis E Discher
- NanoBioPolymers Lab, and Physical Sciences Oncology Center @ Penn , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| |
Collapse
|
24
|
Clarke E, Jarvis CI, Goncalves MB, Kalindjian SB, Adams DR, Brown JT, Shiers JJ, Taddei DM, Ravier E, Barlow S, Miller I, Smith V, Borthwick AD, Corcoran JP. Design and synthesis of a potent, highly selective, orally bioavailable, retinoic acid receptor alpha agonist. Bioorg Med Chem 2018; 26:798-814. [PMID: 29288071 PMCID: PMC5823845 DOI: 10.1016/j.bmc.2017.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/06/2017] [Accepted: 12/08/2017] [Indexed: 11/16/2022]
Abstract
A ligand-based virtual screening exercise examining likely bioactive conformations of AM 580 (2) and AGN 193836 (3) was used to identify the novel, less lipophilic RARα agonist 4-(3,5-dichloro-4-ethoxybenzamido)benzoic acid 5, which has good selectivity over the RARβ, and RARγ receptors. Analysis of the medicinal chemistry parameters of the 3,5-substituents of derivatives of template 5 enabled us to design a class of drug-like molecules with lower intrinsic clearance and higher oral bioavailability which led to the novel RARα agonist 4-(3-chloro-4-ethoxy-5-isopropoxybenzamido)-2-methylbenzoic acid 56 that has high RARα potency and excellent selectivity versus RARβ (2 orders of magnitude) and RARγ (4 orders of magnitude) at both the human and mouse RAR receptors with improved drug-like properties. This RARα specific agonist 56 has high oral bioavailability (>80%) in both mice and dogs with a good PK profile and was shown to be inactive in cytotoxicity and genotoxicity screens.
Collapse
Affiliation(s)
- Earl Clarke
- Neuroscience Drug Discovery Unit, Wolfson Centre for Age-Related Diseases, Guy’s Campus, King’s College, London SE1 1UL, UK
| | - Christopher I. Jarvis
- Neuroscience Drug Discovery Unit, Wolfson Centre for Age-Related Diseases, Guy’s Campus, King’s College, London SE1 1UL, UK
| | - Maria B. Goncalves
- Neuroscience Drug Discovery Unit, Wolfson Centre for Age-Related Diseases, Guy’s Campus, King’s College, London SE1 1UL, UK
| | - S. Barret Kalindjian
- Neuroscience Drug Discovery Unit, Wolfson Centre for Age-Related Diseases, Guy’s Campus, King’s College, London SE1 1UL, UK
| | - David R. Adams
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Jane T. Brown
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Jason J. Shiers
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - David M.A. Taddei
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Elodie Ravier
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Stephanie Barlow
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Iain Miller
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | - Vanessa Smith
- Sygnature Discovery Limited, Biocity, Pennyfoot Street, Nottingham NG1 1GF, UK
| | | | - Jonathan P.T. Corcoran
- Neuroscience Drug Discovery Unit, Wolfson Centre for Age-Related Diseases, Guy’s Campus, King’s College, London SE1 1UL, UK
| |
Collapse
|
25
|
Interactive effects of 9-cis-retinoic acid and androgen on proliferation, differentiation, and apoptosis of LNCaP prostate cancer cells. Eur J Cancer Prev 2018; 26:71-77. [PMID: 26886237 DOI: 10.1097/cej.0000000000000230] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
9-cis-Retinoic acid (9cRA), which binds to both retinoic acid receptors and retinoic X receptors, inhibits prostate cancer induction in rats and reduces growth of prostate cancer cells. However, the nature of this growth inhibition and the interactive influence of androgens are not well defined and are the subject of this report. LNCaP and PC-3 cells were cultured and treated with a range of 9cRA concentrations for 3-6 days in the absence or presence of 5α-dehydrotestosterone. 9cRA inhibited cell proliferation in a dose-dependent manner, plateauing at 10 mol/l. Treatment of cells with 10 mol/l 9cRA inhibited 5α-dihydroxytestosterone (DHT)-stimulated proliferation, the effect of which was maximal at 10 mol/l DHT. Treatment of DHT (10 mol/l)-exposed cells with 9cRA caused a dose-dependent increase in prostate-specific antigen in the medium after 6 days, but not 3 days. 9cRA caused a dose-dependent increase in apoptotic cells stained with H33258 after 3 days, but not 6 days; however, on using flow cytometry, apoptosis was apparent at both 3 and 6 days. Flow cytometry also revealed interference of G0/G1 to S phase transition by 9cRA. Inhibition by 9cRA of anchorage-independent growth of PC-3 cells was also found; LNCaP cells did not grow colonies in soft agar. 9cRA inhibited growth and induced differentiation of human LNCaP prostate cancer cells in vitro and inhibited anchorage-independent growth of PC-3 cells. Because 9cRA and 13-cis-retinoic acid, which is retinoic acid receptor-selective, prevent prostate carcinogenesis in rats, and 13-cis-retinoic acid also inhibits growth of human prostate cancer cells, the RAR is a potential molecular target for prostate cancer prevention and therapy.
Collapse
|
26
|
Yakushiji-Kaminatsui N, Kondo T, Hironaka KI, Sharif J, Endo TA, Nakayama M, Masui O, Koseki Y, Kondo K, Ohara O, Vidal M, Morishita Y, Koseki H. Variant PRC1 competes with retinoic acid-related signals to repress Meis2 in distal forelimb bud. Development 2018; 145:dev.166348. [DOI: 10.1242/dev.166348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/28/2018] [Indexed: 12/12/2022]
Abstract
Suppression of Meis genes in the distal limb bud is required for Proximal-Distal (PD) specification of the forelimb. Polycomb group (PcG) factors play a role in downregulation of retinoic acid (RA)-related signals in the distal forelimb bud, causing Meis repression. It is, however, not known if downregulation of RA-related signals and PcG-mediated proximal genes repression are functionally linked. Here, we reveal that PcG factors and RA-related signals antagonize each other to polarize Meis2 expression along the PD axis. With mathematical modeling and simulation, we propose that PcG factors are required to adjust the threshold for RA-related signaling to regulate Meis2 expression. Finally, we show that a variant Polycomb repressive complex 1 (PRC1), incorporating PCGF3 and PCGF5, represses Meis2 expression in the distal limb bud. Taken together, we reveal a previously unknown link between PcG proteins and downregulation of RA-related signals to mediate the phase transition of Meis2 transcriptional status during forelimb specification.
Collapse
Affiliation(s)
- Nayuta Yakushiji-Kaminatsui
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Takashi Kondo
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- CREST, Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- KAST, Project on Health and Anti-aging, 3-25-13 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Ken-ichi Hironaka
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 113-0033, Japan
| | - Jafar Sharif
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- CREST, Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Takaho A. Endo
- Laboratory for Integrative Genomics, RIKEN-IMS, 1-7-22 Suehirocho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Manabu Nakayama
- Department of Technology Development, Kazusa DNA Research Institute, 2-6-7 Kazusa-Kamatari, Kisarazu, Chiba 292-0818, Japan
| | - Osamu Masui
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- CREST, Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Yoko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- CREST, Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Kaori Kondo
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- CREST, Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- KAST, Project on Health and Anti-aging, 3-25-13 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN-IMS, 1-7-22 Suehirocho, Tsurumi-ku, Yokohama 230-0045, Japan
- Department of Technology Development, Kazusa DNA Research Institute, 2-6-7 Kazusa-Kamatari, Kisarazu, Chiba 292-0818, Japan
| | - Miguel Vidal
- Centro de Investigaciones Biológicas, Department of Cellular and Molecular Biology, Ramiro de Maeztu 9, Madrid 28040, Spain
| | - Yoshihiro Morishita
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
- CREST, Japan Science and Technology Agency, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| |
Collapse
|
27
|
Profiling of the transcriptional response to all-trans retinoic acid in breast cancer cells reveals RARE-independent mechanisms of gene expression. Sci Rep 2017; 7:16684. [PMID: 29192143 PMCID: PMC5709375 DOI: 10.1038/s41598-017-16687-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022] Open
Abstract
Retinoids, derivatives of vitamin A, are key physiological molecules with regulatory effects on cell differentiation, proliferation and apoptosis. As a result, they are of interest for cancer therapy. Specifically, models of breast cancer have varied responses to manipulations of retinoid signaling. This study characterizes the transcriptional response of MDA-MB-231 and MDA-MB-468 breast cancer cells to retinaldehyde dehydrogenase 1A3 (ALDH1A3) and all-trans retinoic acid (atRA). We demonstrate limited overlap between ALDH1A3-induced gene expression and atRA-induced gene expression in both cell lines, suggesting that the function of ALDH1A3 in breast cancer progression extends beyond its role as a retinaldehyde dehydrogenase. Our data reveals divergent transcriptional responses to atRA, which are largely independent of genomic retinoic acid response elements (RAREs) and consistent with the opposing responses of MDA-MB-231 and MDA-MB-468 to in vivo atRA treatment. We identify transcription factors associated with each gene set. Manipulation of the IRF1 transcription factor demonstrates that it is the level of atRA-inducible and epigenetically regulated transcription factors that determine expression of target genes (e.g. CTSS, cathepsin S). This study provides a paradigm for complex responses of breast cancer models to atRA treatment, and illustrates the need to characterize RARE-independent responses to atRA in a variety of models.
Collapse
|
28
|
Chagani S, Wang R, Carpenter EL, Löhr CV, Ganguli-Indra G, Indra AK. Ablation of epidermal RXRα in cooperation with activated CDK4 and oncogenic NRAS generates spontaneous and acute neonatal UVB induced malignant metastatic melanomas. BMC Cancer 2017; 17:736. [PMID: 29121869 PMCID: PMC5679438 DOI: 10.1186/s12885-017-3714-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/30/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Understanding the underlying molecular mechanisms involved in the formation of cutaneous malignant melanoma is critical for improved diagnosis and treatment. Keratinocytic nuclear receptor Retinoid X Receptor α (RXRα) has a protective role against melanomagenesis and is involved in the regulation of keratinocyte and melanocyte homeostasis subsequent acute ultraviolet (UV) irradiation. METHODS We generated a trigenic mouse model system (RXRα ep-/- | Tyr-NRAS Q61K | CDK4 R24C/R24C ) harboring an epidermal knockout of Retinoid X Receptor α (RXRα ep-/- ), combined with oncogenic NRAS Q61K (constitutively active RAS) and activated CDK4 R24C/R24C (constitutively active CDK4). Those mice were subjected to a single neonatal dose of UVB treatment and the role of RXR α was evaluated by characterizing the molecular and cellular changes that took place in the untreated and UVB treated trigenic RXRα ep-/- mice compared to the control mice with functional RXRα. RESULTS Here we report that the trigenic mice develops spontaneous melanoma and exposure to a single neonatal UVB treatment reduces the tumor latency in those mice compared to control mice with functional RXRα. Melanomas from the trigenic RXRα ep-/- mice are substantial in size, show increased proliferation, exhibit increased expression of malignant melanoma markers and exhibit enhanced vascularization. Altered expression of several biomarkers including increased expression of activated AKT, p21 and cyclin D1 and reduced expression of pro-apoptotic marker BAX was observed in the tumor adjacent normal (TAN) skin of acute ultraviolet B treated trigenic RXRα ep-/- mice. Interestingly, we observed a significant increase in p21 and Cyclin D1 in the TAN skin of un-irradiated trigenic RXRα ep-/- mice, suggesting that those changes might be consequences of loss of functional RXRα in the melanoma microenvironment. Loss of RXRα in the epidermal keratinocytes in combination with oncogenic NRAS Q61K and CDK4 R24C/R24C mutations in trigenic mice led to significant melanoma invasion into the draining lymph nodes as compared to controls with functional RXRα. CONCLUSIONS Our study demonstrates the protective role of keratinocytic RxRα in (1) suppressing spontaneous and acute UVB-induced melanoma, and (2) preventing progression of the melanoma to malignancy in the presence of driver mutations like activated CDK4 R24C/R24C and oncogenic NRAS Q61K .
Collapse
Affiliation(s)
- Sharmeen Chagani
- Molecular and Cellular Biology Program, OSU, Corvallis, 97331, OR, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
| | - Rong Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
- Linus Pauling Institute, OSU, Corvallis, OR, USA
| | - Evan L Carpenter
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
| | - Christiane V Löhr
- College of Veterinary Medicine, Oregon State University, Corvallis, Oregon, 97331, USA
| | - Gitali Ganguli-Indra
- Molecular and Cellular Biology Program, OSU, Corvallis, 97331, OR, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, 97239, OR, USA
| | - Arup K Indra
- Molecular and Cellular Biology Program, OSU, Corvallis, 97331, OR, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, OSU, Corvallis, 97331, OR, USA.
- Linus Pauling Institute, OSU, Corvallis, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University (OHSU), Portland, 97239, OR, USA.
- Department of Dermatology, OHSU, Portland, 97239, OR, USA.
| |
Collapse
|
29
|
Chandra V, Wu D, Li S, Potluri N, Kim Y, Rastinejad F. The quaternary architecture of RARβ-RXRα heterodimer facilitates domain-domain signal transmission. Nat Commun 2017; 8:868. [PMID: 29021580 PMCID: PMC5636793 DOI: 10.1038/s41467-017-00981-y] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
Assessing the physical connections and allosteric communications in multi-domain nuclear receptor (NR) polypeptides has remained challenging, with few crystal structures available to show their overall structural organizations. Here we report the quaternary architecture of multi-domain retinoic acid receptor β-retinoic X receptor α (RARβ-RXRα) heterodimer bound to DNA, ligands and coactivator peptides, examined through crystallographic, hydrogen-deuterium exchange mass spectrometry, mutagenesis and functional studies. The RARβ ligand-binding domain (LBD) and DNA-binding domain (DBD) are physically connected to foster allosteric signal transmission between them. Direct comparisons among all the multi-domain NRs studied crystallographically to date show significant variations within their quaternary architectures, rather than a common architecture adhering to strict rules. RXR remains flexible and adaptive by maintaining loosely organized domains, while its heterodimerization partners use a surface patch on their LBDs to form domain-domain interactions with DBDs.Nuclear receptors (NR) are multidomain proteins, which makes their crystallization challenging. Here the authors present the crystal structure of the retinoic acid receptor β-retinoic X receptor α (RARβ-RXRα) heterodimer bound to DNA, ligands and coactivator peptides, which shows that NR quaternary architectures are variable.
Collapse
Affiliation(s)
- Vikas Chandra
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, 32827, USA
| | - Dalei Wu
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, 32827, USA
- Shandong University-Helmholtz Institute of Biotechnology, State Key Laboratory of Microbial Technology, School of Life Sciences, Shandong University, Qingdao, Shandong, 266237, China
| | - Sheng Li
- Department of Medicine and UCSD DXMS Proteomics Resource, University of California, San Diego, La Jolla, CA, 92023, USA
| | - Nalini Potluri
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, 32827, USA
| | - Youngchang Kim
- Structural Biology Center, Biosciences Division, Argonne National Laboratory, Argonne, IL, 60439, USA
| | - Fraydoon Rastinejad
- Integrative Metabolism Program, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL, 32827, USA.
| |
Collapse
|
30
|
Bruschi FV, Claudel T, Tardelli M, Caligiuri A, Stulnig TM, Marra F, Trauner M. The PNPLA3 I148M variant modulates the fibrogenic phenotype of human hepatic stellate cells. Hepatology 2017; 65:1875-1890. [PMID: 28073161 DOI: 10.1002/hep.29041] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/23/2016] [Indexed: 12/12/2022]
Abstract
UNLABELLED The genetic polymorphism I148M of patatin-like phospholipase domain-containing 3 (PNPLA3) is robustly associated with hepatic steatosis and its progression to steatohepatitis, fibrosis, and cancer. Hepatic stellate cells (HSCs) are key players in the development of liver fibrosis, but the role of PNPLA3 and its variant I148M in this process is poorly understood. Here we analyzed the expression of PNPLA3 during human HSC activation and thereby explored how a PNPLA3 variant impacts hepatic fibrogenesis. We show that expression of PNPLA3 gene and protein increases during the early phases of activation and remains elevated in fully activated HSCs (P < 0.01). Knockdown of PNPLA3 significantly decreases the profibrogenic protein alpha-smooth muscle actin (P < 0.05). Primary human I148M HSCs displayed significantly higher expression and release of proinflammatory cytokines, such as chemokine (C-C motif) ligand 5 (P < 0.01) and granulocyte-macrophage colony-stimulating factor (P < 0.001), thus contributing to migration of immune cells (P < 0.05). Primary I148M HSCs showed reduced retinol (P < 0.001) but higher lipid droplet content (P < 0.001). In line with this, LX-2 cells stably overexpressing I148M showed augmented proliferation and migration, lower retinol, and abolished retinoid X receptor/retinoid A receptor transcriptional activities but more lipid droplets. Knockdown of I148M PNPLA3 (P < 0.001) also reduces chemokine (C-C motif) ligand 5 and collagen1α1 expression (P < 0.05). Notably, I148M cells display reduced peroxisome proliferator-activated receptor gamma transcriptional activity, and this effect was attributed to increased c-Jun N-terminal kinase, thereby inhibiting peroxisome proliferator-activated receptor gamma through serine 84 phosphorylation and promoting activator protein 1 transcription. Conversely, the c-Jun N-terminal kinase inhibitor SP600125 and the peroxisome proliferator-activated receptor gamma agonist rosiglitazone decreased activator protein 1 promoter activity. CONCLUSIONS These data indicate that PNPLA3 is required for HSC activation and that its genetic variant I148M potentiates the profibrogenic features of HSCs, providing a molecular mechanism for the higher risk of progression and severity of liver diseases conferred to patients carrying the I148M variant. (Hepatology 2017;65:1875-1890).
Collapse
Affiliation(s)
- Francesca Virginia Bruschi
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Medical University of Vienna, Vienna, Austria
| | - Thierry Claudel
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Medical University of Vienna, Vienna, Austria
| | - Matteo Tardelli
- Christian Doppler-Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Alessandra Caligiuri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Thomas M Stulnig
- Christian Doppler-Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Fabio Marra
- Christian Doppler-Laboratory for Cardio-Metabolic Immunotherapy and Clinical Division of Endocrinology and Metabolism, Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Hans Popper Laboratory of Molecular Hepatology, Division of Gastroenterology & Hepatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
31
|
Dixon K, Chen J, Li Q. Gene expression profiling discerns molecular pathways elicited by ligand signaling to enhance the specification of embryonic stem cells into skeletal muscle lineage. Cell Biosci 2017; 7:23. [PMID: 28469839 PMCID: PMC5414197 DOI: 10.1186/s13578-017-0150-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/26/2017] [Indexed: 01/07/2023] Open
Abstract
Regulation of lineage specification and differentiation in embryonic stem (ES) cells can be achieved through the activation of endogenous signaling, an avenue for potential application in regenerative medicine. During vertebrate development, retinoic acid (RA) plays an important role in body axis elongation and mesoderm segmentation in that graded exposure to RA provides cells with positional identity and directs commitment to specific tissue lineages. Nevertheless, bexarotene, a clinically approved rexinoid, enhances the specification and differentiation of ES cells into skeletal myocytes more effectively than RA. Thus profiling the transcriptomes of ES cells differentiated with bexarotene or RA permits the identification of different genetic targets and signaling pathways that may contribute to the difference of bexarotene and RA in efficiency of myogenesis. Interestingly, bexarotene induces the early expression of a myogenic progenitor marker, Meox1, while the expression of many RA targets is also enhanced by bexarotene. Several signaling molecules involved in the progression of myogenic specification and commitment are differentially regulated by bexarotene and RA, suggesting that early targets of rexinoid allow the coordinated regulation of molecular events which leads to efficient myogenic differentiation in ES cells.
Collapse
Affiliation(s)
- Katherine Dixon
- 0000 0001 2182 2255grid.28046.38Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2537, Ottawa, ON K1H 8M5 Canada
| | - Jihong Chen
- 0000 0001 2182 2255grid.28046.38Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON Canada
| | - Qiao Li
- 0000 0001 2182 2255grid.28046.38Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Room 2537, Ottawa, ON K1H 8M5 Canada ,0000 0001 2182 2255grid.28046.38Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON Canada
| |
Collapse
|
32
|
Ables ET, Drummond-Barbosa D. Steroid Hormones and the Physiological Regulation of Tissue-Resident Stem Cells: Lessons from the Drosophila Ovary. CURRENT STEM CELL REPORTS 2017; 3:9-18. [PMID: 28458991 PMCID: PMC5407287 DOI: 10.1007/s40778-017-0070-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Stem cells respond to local paracrine signals; more recently, however, systemic hormones have also emerged as key regulators of stem cells. This review explores the role of steroid hormones in stem cells, using the Drosophila germline stem cell as a centerpiece for discussion. RECENT FINDINGS Stem cells sense and respond directly and indirectly to steroid hormones, which regulate diverse sets of target genes via interactions with nuclear hormone receptors. Hormone-regulated networks likely integrate the actions of multiple systemic signals to adjust the activity of stem cell lineages in response to changes in physiological status. SUMMARY Hormones are inextricably linked to animal physiology, and can control stem cells and their local niches. Elucidating the molecular mechanisms of hormone signaling in stem cells is essential for our understanding of the fundamental underpinnings of stem cell biology, and for informing new therapeutic interventions against cancers or for regenerative medicine.
Collapse
Affiliation(s)
- Elizabeth T. Ables
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
33
|
Haffez H, Chisholm DR, Valentine R, Pohl E, Redfern C, Whiting A. The molecular basis of the interactions between synthetic retinoic acid analogues and the retinoic acid receptors. MEDCHEMCOMM 2017; 8:578-592. [PMID: 30108774 PMCID: PMC6072416 DOI: 10.1039/c6md00680a] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/17/2017] [Indexed: 01/17/2023]
Abstract
All-trans-retinoic acid (ATRA) and its synthetic analogues EC23 and EC19 direct cellular differentiation by interacting as ligands for the retinoic acid receptor (RARα, β and γ) family of nuclear receptor proteins. To date, a number of crystal structures of natural and synthetic ligands complexed to their target proteins have been solved, providing molecular level snap-shots of ligand binding. However, a deeper understanding of receptor and ligand flexibility and conformational freedom is required to develop stable and effective ATRA analogues for clinical use. Therefore, we have used molecular modelling techniques to define RAR interactions with ATRA and two synthetic analogues, EC19 and EC23, and compared their predicted biochemical activities to experimental measurements of relative ligand affinity and recruitment of coactivator proteins. A comprehensive molecular docking approach that explored the conformational space of the ligands indicated that ATRA is able to bind the three RAR proteins in a number of conformations with one extended structure being favoured. In contrast the biologically-distinct isomer, 9-cis-retinoic acid (; 9CRA), showed significantly less conformational flexibility in the RAR binding pockets. These findings were used to inform docking studies of the synthetic retinoids EC23 and EC19, and their respective methyl esters. EC23 was found to be an excellent mimic for ATRA, and occupied similar binding modes to ATRA in all three target RAR proteins. In comparison, EC19 exhibited an alternative binding mode which reduces the strength of key polar interactions in RARα/γ but is well-suited to the larger RARβ binding pocket. In contrast, docking of the corresponding esters revealed the loss of key polar interactions which may explain the much reduced biological activity. Our computational results were complemented using an in vitro binding assay based on FRET measurements, which showed that EC23 was a strongly binding, pan-agonist of the RARs, while EC19 exhibited specificity for RARβ, as predicted by the docking studies. These findings can account for the distinct behaviour of EC23 and EC19 in cellular differentiation assays, and additionally, the methods described herein can be further applied to the understanding of the molecular basis for the selectivity of different retinoids to RARα, β and γ.
Collapse
Affiliation(s)
- Hesham Haffez
- Department of Chemistry Durham University , South Road , Durham , DH1 3LE , UK .
- Department of Biosciences & Biophysical Sciences , Institute Durham University , South Road , Durham DH1 3LE , UK
- Northern Institute for Cancer Research , Medical School , Newcastle University , Newcastle upon Tyne , NE2 4HH , UK
- Department of Biochemistry and Molecular Biology , Pharmacy College , Helwan University , Cairo , Egypt
| | - David R Chisholm
- Department of Chemistry Durham University , South Road , Durham , DH1 3LE , UK .
| | - Roy Valentine
- High Force Research Ltd. , Bowburn North Industrial Estate , Bowburn , Durham , DH6 5PF , UK
| | - Ehmke Pohl
- Department of Biosciences & Biophysical Sciences , Institute Durham University , South Road , Durham DH1 3LE , UK
| | - Christopher Redfern
- Northern Institute for Cancer Research , Medical School , Newcastle University , Newcastle upon Tyne , NE2 4HH , UK
| | - Andrew Whiting
- Department of Chemistry Durham University , South Road , Durham , DH1 3LE , UK .
| |
Collapse
|
34
|
A High Throughput Phenotypic Screening reveals compounds that counteract premature osteogenic differentiation of HGPS iPS-derived mesenchymal stem cells. Sci Rep 2016; 6:34798. [PMID: 27739443 PMCID: PMC5064407 DOI: 10.1038/srep34798] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/12/2016] [Indexed: 12/19/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare fatal genetic disorder that causes systemic accelerated aging in children. Thanks to the pluripotency and self-renewal properties of induced pluripotent stem cells (iPSC), HGPS iPSC-based modeling opens up the possibility of access to different relevant cell types for pharmacological approaches. In this study, 2800 small molecules were explored using high-throughput screening, looking for compounds that could potentially reduce the alkaline phosphatase activity of HGPS mesenchymal stem cells (MSCs) committed into osteogenic differentiation. Results revealed seven compounds that normalized the osteogenic differentiation process and, among these, all-trans retinoic acid and 13-cis-retinoic acid, that also decreased progerin expression. This study highlights the potential of high-throughput drug screening using HGPS iPS-derived cells, in order to find therapeutic compounds for HGPS and, potentially, for other aging-related disorders.
Collapse
|
35
|
Comptour A, Rouzaire M, Belville C, Bouvier D, Gallot D, Blanchon L, Sapin V. Nuclear retinoid receptors and pregnancy: placental transfer, functions, and pharmacological aspects. Cell Mol Life Sci 2016; 73:3823-37. [PMID: 27502420 PMCID: PMC11108506 DOI: 10.1007/s00018-016-2332-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 10/21/2022]
Abstract
Animal models of vitamin A (retinol) deficiency have highlighted its crucial role in reproduction and placentation, whereas an excess of retinoids (structurally or functionally related entities) can cause toxic and teratogenic effects in the embryo and foetus, especially in the first trimester of human pregnancy. Knock-out experimental strategies-targeting retinoid nuclear receptors RARs and RXRs have confirmed that the effects of vitamin A are mediated by retinoic acid (especially all-trans retinoic acid) and that this vitamin is essential for the developmental process. All these data show that the vitamin A pathway and metabolism are as important for the well-being of the foetus, as they are for that of the adult. Accordingly, during this last decade, extensive research on retinoid metabolism has yielded detailed knowledge on all the actors in this pathway, spurring the development of antagonists and agonists for therapeutic and research applications. Natural and synthetic retinoids are currently used in clinical practice, most often on the skin for the treatment of acne, and as anti-oncogenic agents in acute promyelocytic leukaemia. However, because of the toxicity and teratogenicity of retinoids during pregnancy, their pharmacological use needs a sound knowledge of their metabolism, molecular aspects, placental transfer, and action.
Collapse
Affiliation(s)
- Aurélie Comptour
- EA7281, Retinoids, Reproduction Developmental Diseases, School of Medicine, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France
| | - Marion Rouzaire
- EA7281, Retinoids, Reproduction Developmental Diseases, School of Medicine, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France
| | - Corinne Belville
- EA7281, Retinoids, Reproduction Developmental Diseases, School of Medicine, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France
- GReD, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France
| | - Damien Bouvier
- EA7281, Retinoids, Reproduction Developmental Diseases, School of Medicine, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France
- Biochemistry and Molecular Biology Department, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Denis Gallot
- EA7281, Retinoids, Reproduction Developmental Diseases, School of Medicine, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France
- Obstetrics and Gynecology Department, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Loïc Blanchon
- EA7281, Retinoids, Reproduction Developmental Diseases, School of Medicine, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France
| | - Vincent Sapin
- EA7281, Retinoids, Reproduction Developmental Diseases, School of Medicine, Clermont Université, Université d'Auvergne, 63000, Clermont-Ferrand, France.
- Biochemistry and Molecular Biology Department, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France.
- Laboratoire de Biochimie Médicale, 4R3, Faculté de Médecine, 28 Place Henri-Dunant, BP38, 63001, Clermont-Ferrand Cedex, France.
| |
Collapse
|
36
|
Heck MC, Wagner CE, Shahani PH, MacNeill M, Grozic A, Darwaiz T, Shimabuku M, Deans DG, Robinson NM, Salama SH, Ziller JW, Ma N, van der Vaart A, Marshall PA, Jurutka PW. Modeling, Synthesis, and Biological Evaluation of Potential Retinoid X Receptor (RXR)-Selective Agonists: Analogues of 4-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethynyl]benzoic Acid (Bexarotene) and 6-(Ethyl(5,5,8,8-tetrahydronaphthalen-2-yl)amino)nicotinic Acid (NEt-TMN). J Med Chem 2016; 59:8924-8940. [DOI: 10.1021/acs.jmedchem.6b00812] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Michael C. Heck
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Carl E. Wagner
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Pritika H. Shahani
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Mairi MacNeill
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Aleksandra Grozic
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Tamana Darwaiz
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Micah Shimabuku
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - David G. Deans
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Nathan M. Robinson
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Samer H. Salama
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Joseph W. Ziller
- Department
of Chemistry, University of California, Irvine, 576 Rowland Hall, Irvine, California 92697, United States
| | - Ning Ma
- Department
of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, United States
| | - Arjan van der Vaart
- Department
of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida 33620, United States
| | - Pamela A. Marshall
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| | - Peter W. Jurutka
- School
of Mathematical and Natural Sciences, New College of Interdisciplinary
Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States
| |
Collapse
|
37
|
Zhu S, Guleria RS, Thomas CM, Roth A, Gerilechaogetu F, Kumar R, Dostal DE, Baker KM, Pan J. Loss of myocardial retinoic acid receptor α induces diastolic dysfunction by promoting intracellular oxidative stress and calcium mishandling in adult mice. J Mol Cell Cardiol 2016; 99:100-112. [PMID: 27539860 DOI: 10.1016/j.yjmcc.2016.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/10/2016] [Accepted: 08/12/2016] [Indexed: 01/09/2023]
Abstract
Retinoic acid receptor (RAR) has been implicated in pathological stimuli-induced cardiac remodeling. To determine whether the impairment of RARα signaling directly contributes to the development of heart dysfunction and the involved mechanisms, tamoxifen-induced myocardial specific RARα deletion (RARαKO) mice were utilized. Echocardiographic and cardiac catheterization studies showed significant diastolic dysfunction after 16wks of gene deletion. However, no significant differences were observed in left ventricular ejection fraction (LVEF), between RARαKO and wild type (WT) control mice. DHE staining showed increased intracellular reactive oxygen species (ROS) generation in the hearts of RARαKO mice. Significantly increased NOX2 (NADPH oxidase 2) and NOX4 levels and decreased SOD1 and SOD2 levels were observed in RARαKO mouse hearts, which were rescued by overexpression of RARα in cardiomyocytes. Decreased SERCA2a expression and phosphorylation of phospholamban (PLB), along with decreased phosphorylation of Akt and Ca2+/calmodulin-dependent protein kinase II δ (CaMKII δ) was observed in RARαKO mouse hearts. Ca2+ reuptake and cardiomyocyte relaxation were delayed by RARα deletion. Overexpression of RARα or inhibition of ROS generation or NOX activation prevented RARα deletion-induced decrease in SERCA2a expression/activation and delayed Ca2+ reuptake. Moreover, the gene and protein expression of RARα was significantly decreased in aged or metabolic stressed mouse hearts. RARα deletion accelerated the development of diastolic dysfunction in streptozotocin (STZ)-induced type 1 diabetic mice or in high fat diet fed mice. In conclusion, myocardial RARα deletion promoted diastolic dysfunction, with a relative preserved LVEF. Increased oxidative stress have an important role in the decreased expression/activation of SERCA2a and Ca2+ mishandling in RARαKO mice, which are major contributing factors in the development of diastolic dysfunction. These data suggest that impairment of cardiac RARα signaling may be a novel mechanism that is directly linked to pathological stimuli-induced diastolic dysfunction.
Collapse
Affiliation(s)
- Sen Zhu
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Rakeshwar S Guleria
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States.
| | - Candice M Thomas
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Amanda Roth
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Fnu Gerilechaogetu
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Rajesh Kumar
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - David E Dostal
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Kenneth M Baker
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States
| | - Jing Pan
- Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States; Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Central Texas Veterans Health Care System, Baylor Scott & White Health, Temple, TX, United States.
| |
Collapse
|
38
|
Olivares AM, Moreno-Ramos OA, Haider NB. Role of Nuclear Receptors in Central Nervous System Development and Associated Diseases. J Exp Neurosci 2016; 9:93-121. [PMID: 27168725 PMCID: PMC4859451 DOI: 10.4137/jen.s25480] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 11/13/2022] Open
Abstract
The nuclear hormone receptor (NHR) superfamily is composed of a wide range of receptors involved in a myriad of important biological processes, including development, growth, metabolism, and maintenance. Regulation of such wide variety of functions requires a complex system of gene regulation that includes interaction with transcription factors, chromatin-modifying complex, and the proper recognition of ligands. NHRs are able to coordinate the expression of genes in numerous pathways simultaneously. This review focuses on the role of nuclear receptors in the central nervous system and, in particular, their role in regulating the proper development and function of the brain and the eye. In addition, the review highlights the impact of mutations in NHRs on a spectrum of human diseases from autism to retinal degeneration.
Collapse
Affiliation(s)
- Ana Maria Olivares
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Oscar Andrés Moreno-Ramos
- Departamento de Ciencias Biológicas, Facultad de Ciencias, Universidad de los Andes, Bogotá, Colombia
| | - Neena B Haider
- Department of Ophthalmology, Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Chen J, Li Q. Implication of retinoic acid receptor selective signaling in myogenic differentiation. Sci Rep 2016; 6:18856. [PMID: 26830006 PMCID: PMC4735650 DOI: 10.1038/srep18856] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/25/2015] [Indexed: 11/18/2022] Open
Abstract
Signaling molecules are important for committing individual cells into tissue-specific lineages during early vertebrate development. Retinoic acid (RA) is an important vertebrate morphogen, in that its concentration gradient is essential for correct patterning of the vertebrate embryo. RA signaling is mediated through the activation of retinoic acid receptors (RARs), which function as ligand-dependent transcription factors. In this study, we examined the molecular mechanisms of RAR-selective signaling in myogenic differentiation. We found that just like natural ligand RA, a RAR-selective ligand is an effective enhancer in the commitment of skeletal muscle lineage at the early stage of myogenic differentiation. Interestingly, the kinetics and molecular basis of the RAR-selective ligand in myogenic differentiation are similar to that of natural ligand RA. Also similar to natural ligand RA, the RAR-selective ligand enhances myogenic differentiation through β-catenin signaling pathway while inhibiting cardiac differentiation. Furthermore, while low concentrations of natural ligand RA or RAR-selective ligand regulate myogenic differentiation through RAR function and coactivator recruitment, high concentrations are critical to the expression of a model RA-responsive gene. Thus our data suggests that RAR-mediated gene regulation may be highly context-dependent, affected by locus-specific interaction or local chromatin environment.
Collapse
Affiliation(s)
- Jihong Chen
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Qiao Li
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.,Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
40
|
Hurwitz JL, Penkert RR, Xu B, Fan Y, Partridge JF, Maul RW, Gearhart PJ. Hotspots for Vitamin-Steroid-Thyroid Hormone Response Elements Within Switch Regions of Immunoglobulin Heavy Chain Loci Predict a Direct Influence of Vitamins and Hormones on B Cell Class Switch Recombination. Viral Immunol 2016; 29:132-6. [PMID: 26741514 DOI: 10.1089/vim.2015.0104] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Vitamin A deficiencies are common throughout the world and have a significant negative influence on immune protection against viral infections. Mouse models demonstrate that the production of IgA, a first line of defense against viruses at mucosal sites, is inhibited in the context of vitamin A deficiency. In vitro, the addition of vitamin A to activated B cells can enhance IgA expression, but downregulate IgE. Previous reports have demonstrated that vitamin A modifies cytokine patterns, and in so doing may influence antibody isotype expression by an indirect mechanism. However, we have now discovered hundreds of potential response elements among Sμ, Sɛ, and Sα switch sites within immunoglobulin heavy chain loci. These hotspots appear in both mouse and human loci and include targets for vitamin receptors and related proteins (e.g., estrogen receptors) in the nuclear receptor superfamily. Full response elements with direct repeats are relatively infrequent or absent in Sγ regions although half-sites are present. Based on these results, we pose a hypothesis that nuclear receptors have a direct effect on the immunoglobulin heavy chain class switch recombination event. We propose that vitamin A may alter S site accessibility to activation-induced deaminase and nonhomologous end-joining machinery, thereby influencing the isotype switch, antibody production, and protection against viral infections at mucosal sites.
Collapse
Affiliation(s)
- Julia L Hurwitz
- 1 Department of Infectious Diseases, St. Jude Children's Research Hospital , Memphis, Tennessee.,2 Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center , Memphis, Tennessee
| | - Rhiannon R Penkert
- 1 Department of Infectious Diseases, St. Jude Children's Research Hospital , Memphis, Tennessee
| | - Beisi Xu
- 3 Department of Computational Biology, St. Jude Children's Research Hospital , Memphis, Tennessee
| | - Yiping Fan
- 3 Department of Computational Biology, St. Jude Children's Research Hospital , Memphis, Tennessee
| | - Janet F Partridge
- 4 Department of Pathology, St. Jude Children's Research Hospital , Memphis, Tennessee
| | - Robert W Maul
- 5 National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| | - Patricia J Gearhart
- 5 National Institute on Aging, National Institutes of Health , Baltimore, Maryland
| |
Collapse
|
41
|
Green AC, Martin TJ, Purton LE. The role of vitamin A and retinoic acid receptor signaling in post-natal maintenance of bone. J Steroid Biochem Mol Biol 2016; 155:135-46. [PMID: 26435449 DOI: 10.1016/j.jsbmb.2015.09.036] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 09/24/2015] [Accepted: 09/26/2015] [Indexed: 12/11/2022]
Abstract
Vitamin A and retinoid derivatives are recognized as morphogens that govern body patterning and skeletogenesis, producing profound defects when in excess. In post-natal bone, both high and low levels of vitamin A are associated with poor bone heath and elevated risk of fractures. Despite this, the precise mechanism of how retinoids induce post-natal bone changes remains elusive. Numerous studies have been performed to discover how retinoids induce these changes, revealing a complex morphogenic regulation of bone through interplay of different cell types. This review will discuss the direct and indirect effects of retinoids on mediators of bone turnover focusing on differentiation and activity of osteoblasts and osteoclasts and explains why some discrepancies in this field have arisen. Importantly, the overall effect of retinoids on the skeleton is highly site-specific, likely due to differential regulation of osteoblasts and osteoclasts at trabecular vs. cortical periosteal and endosteal bone surfaces. Further investigation is required to discover the direct gene targets of retinoic acid receptors (RARs) and molecular mechanisms through which these changes occur. A clear role for RARs in regulating bone is now accepted and the therapeutic potential of retinoids in treating bone diseases has been established.
Collapse
Affiliation(s)
- Alanna C Green
- St Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia.
| | - T John Martin
- St Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia
| | - Louise E Purton
- St Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia
| |
Collapse
|
42
|
AlSudais H, Aabed K, Nicola W, Dixon K, Chen J, Li Q. Retinoid X Receptor-selective Signaling in the Regulation of Akt/Protein Kinase B Isoform-specific Expression. J Biol Chem 2015; 291:3090-9. [PMID: 26668312 DOI: 10.1074/jbc.m115.692707] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Indexed: 01/22/2023] Open
Abstract
The differentiation and fusion of myoblasts into mature myotubes are complex processes responding to multiple signaling pathways. The function of Akt/PKB is critical for myogenesis, but less is clear as to the regulation of its isoform-specific expression. Bexarotene is a drug already used clinically to treat cancer, and it has the ability to enhance the commitment of embryonic stem cells into skeletal muscle lineage. Whereas bexarotene regulates fundamental biological processes through retinoid X receptor (RXR)-mediated gene expression, molecular pathways underlying its positive effects on myogenesis remain unclear. In this study, we have examined the signaling pathways that transmit bexarotene action in the context of myoblast differentiation. We show that bexarotene promotes myoblast differentiation and fusion through the activation of RXR and the regulation of Akt/PKB isoform-specific expression. Interestingly, bexarotene signaling appears to correlate with residue-specific histone acetylation and is able to counteract the detrimental effects of cachectic factors on myogenic differentiation. We also signify an isoform-specific role for Akt/PKB in RXR-selective signaling to promote and to retain myoblast differentiation. Taken together, our findings establish the viability of applying bexarotene in the prevention and treatment of muscle-wasting disorders, particularly given the lack of drugs that promote myogenic differentiation available for potential clinical applications. Furthermore, the model of bexarotene-enhanced myogenic differentiation will provide an important avenue to identify additional genetic targets and specific molecular interactions that we can study and apply for the development of potential therapeutics in muscle regeneration and repair.
Collapse
Affiliation(s)
| | - Kawther Aabed
- Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - William Nicola
- From the Departments of Cellular and Molecular Medicine and
| | | | - Jihong Chen
- Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Qiao Li
- From the Departments of Cellular and Molecular Medicine and Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
43
|
Stoney PN, Helfer G, Rodrigues D, Morgan PJ, McCaffery P. Thyroid hormone activation of retinoic acid synthesis in hypothalamic tanycytes. Glia 2015; 64:425-39. [PMID: 26527258 PMCID: PMC4949630 DOI: 10.1002/glia.22938] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 10/08/2015] [Accepted: 10/12/2015] [Indexed: 11/11/2022]
Abstract
Thyroid hormone (TH) is essential for adult brain function and its actions include several key roles in the hypothalamus. Although TH controls gene expression via specific TH receptors of the nuclear receptor class, surprisingly few genes have been demonstrated to be directly regulated by TH in the hypothalamus, or the adult brain as a whole. This study explored the rapid induction by TH of retinaldehyde dehydrogenase 1 (Raldh1), encoding a retinoic acid (RA)-synthesizing enzyme, as a gene specifically expressed in hypothalamic tanycytes, cells that mediate a number of actions of TH in the hypothalamus. The resulting increase in RA may then regulate gene expression via the RA receptors, also of the nuclear receptor class. In vivo exposure of the rat to TH led to a significant and rapid increase in hypothalamic Raldh1 within 4 hours. That this may lead to an in vivo increase in RA is suggested by the later induction by TH of the RA-responsive gene Cyp26b1. To explore the actions of RA in the hypothalamus as a potential mediator of TH control of gene regulation, an ex vivo hypothalamic rat slice culture method was developed in which the Raldh1-expressing tanycytes were maintained. These slice cultures confirmed that TH did not act on genes regulating energy balance but could induce Raldh1. RA has the potential to upregulate expression of genes involved in growth and appetite, Ghrh and Agrp. This regulation is acutely sensitive to epigenetic changes, as has been shown for TH action in vivo. These results indicate that sequential triggering of two nuclear receptor signalling systems has the capability to mediate some of the functions of TH in the hypothalamus.
Collapse
Affiliation(s)
- Patrick N Stoney
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, AB25 2ZD, United Kingdom
| | - Gisela Helfer
- Rowett Institute of Nutrition and Health, University of Aberdeen, Bucksburn, Aberdeen, Scotland, AB21 9SB, United Kingdom
| | - Diana Rodrigues
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, AB25 2ZD, United Kingdom
| | - Peter J Morgan
- Rowett Institute of Nutrition and Health, University of Aberdeen, Bucksburn, Aberdeen, Scotland, AB21 9SB, United Kingdom
| | - Peter McCaffery
- School of Medical Sciences, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, Scotland, AB25 2ZD, United Kingdom
| |
Collapse
|
44
|
Chen HJ, Chiang BL. Effect of Hyperoxia on Retinoid Metabolism and Retinoid Receptor Expression in the Lungs of Newborn Mice. PLoS One 2015; 10:e0140343. [PMID: 26509921 PMCID: PMC4624932 DOI: 10.1371/journal.pone.0140343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 09/24/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Preterm newborns that receive oxygen therapy often develop bronchopulmonary dysplasia (BPD), which is abnormal lung development characterized by impaired alveologenesis. Oxygen-mediated injury is thought to disrupt normal lung growth and development. However, the mechanism of hyperoxia-induced BPD has not been extensively investigated. We established a neonatal mouse model to investigate the effects of normobaric hyperoxia on retinoid metabolism and retinoid receptor expression. METHODS Newborn mice were exposed to hyperoxic or normoxic conditions for 15 days. The concentration of retinol and retinyl palmitate in the lung was measured by HPLC to gauge retinoid metabolism. Retinoid receptor mRNA levels were assessed by real-time PCR. Proliferation and retinoid receptor expression in A549 cells were assessed in the presence and absence of exogenous vitamin A. RESULTS Hyperoxia significantly reduced the body and lung weight of neonatal mice. Hyperoxia also downregulated expression of RARα, RARγ, and RXRγ in the lungs of neonatal mice. In vitro, hyperoxia inhibited proliferation and expression of retinoid receptors in A549 cells. CONCLUSION Hyperoxia disrupted retinoid receptor expression in neonatal mice.
Collapse
Affiliation(s)
- Hsing-Jin Chen
- Graduate Institute of Clinical Medicine College of Medicine of National Taiwan University, Taipei, Taiwan
| | - Bor-Luen Chiang
- Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
45
|
Marshall PA, Jurutka PW, Wagner CE, van der Vaart A, Kaneko I, Chavez PI, Ma N, Bhogal JS, Shahani P, Swierski JC, MacNeill M. Analysis of differential secondary effects of novel rexinoids: select rexinoid X receptor ligands demonstrate differentiated side effect profiles. Pharmacol Res Perspect 2015; 3:e00122. [PMID: 26038698 PMCID: PMC4448986 DOI: 10.1002/prp2.122] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/28/2014] [Accepted: 12/15/2014] [Indexed: 11/21/2022] Open
Abstract
In order to determine the feasibility of utilizing novel rexinoids for chemotherapeutics and as potential treatments for neurological conditions, we undertook an assessment of the side effect profile of select rexinoid X receptor (RXR) analogs that we reported previously. We assessed pharmacokinetic profiles, lipid and thyroid-stimulating hormone (TSH) levels in rats, and cell culture activity of rexinoids in sterol regulatory element-binding protein (SREBP) induction and thyroid hormone inhibition assays. We also performed RNA sequencing of the brain tissues of rats that had been dosed with the compounds. We show here for the first time that potent rexinoid activity can be uncoupled from drastic lipid changes and thyroid axis variations, and we propose that rexinoids can be developed with improved side effect profiles than the parent compound, bexarotene (1).
Collapse
Affiliation(s)
- Pamela A Marshall
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| | - Peter W Jurutka
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| | - Carl E Wagner
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| | - Arjan van der Vaart
- Department of Chemistry, University of South Florida 4202 E Fowler Ave CHE 205, Tampa, Florida, 33620
| | - Ichiro Kaneko
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| | - Pedro I Chavez
- Biomedical Sciences Program, Midwestern University 19555 N 59th Ave., Glendale, Arizona, 86308
| | - Ning Ma
- Department of Chemistry, University of South Florida 4202 E Fowler Ave CHE 205, Tampa, Florida, 33620
| | - Jaskaran S Bhogal
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| | - Pritika Shahani
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| | - Johnathon C Swierski
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| | - Mairi MacNeill
- School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University 4701 W Thunderbird Rd, Glendale, Arizona, 85306
| |
Collapse
|
46
|
Boku S, Toda H, Nakagawa S, Kato A, Inoue T, Koyama T, Hiroi N, Kusumi I. Neonatal maternal separation alters the capacity of adult neural precursor cells to differentiate into neurons via methylation of retinoic acid receptor gene promoter. Biol Psychiatry 2015; 77:335-44. [PMID: 25127741 PMCID: PMC5241093 DOI: 10.1016/j.biopsych.2014.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 07/05/2014] [Accepted: 07/09/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Early life stress is thought to contribute to psychiatric disorders, but the precise mechanisms underlying this link are poorly understood. As neonatal stress decreases adult hippocampal neurogenesis, which, in turn, functionally contributes to many behavioral phenotypes relevant to psychiatric disorders, we examined how in vivo neonatal maternal separation (NMS) impacts the capacity of adult hippocampal neural precursor cells via epigenetic alterations in vitro. METHODS Rat pups were separated from their dams for 3 hours daily from postnatal day (PND) 2 to PND 14 or were never separated from the dam (as control animals). We isolated adult neural precursor cells from the hippocampal dentate gyrus at PND 56 and assessed rates of proliferation, apoptosis, and differentiation in cell culture. We also evaluated the effect of DNA methylation at the retinoic acid receptor (RAR) promoter stemming from NMS on adult neural precursor cells. RESULTS NMS attenuated neural differentiation of adult neural precursor cells but had no detectible effect on proliferation, apoptosis, or astroglial differentiation. The DNA methyltransferase (DNMT) inhibitor, 5-aza-dC, reversed a reduction by NMS of neural differentiation of adult neural precursor cells. NMS increased DNMT1 expression and decreased expression of RARα. An RARα agonist increased neural differentiation and an antagonist reduced retinoic acid-induced neural differentiation. NMS increased the methylated portion of RARα promoter, and the DNMT inhibitor reversed a reduction by NMS of RARα messenger RNA expression. CONCLUSIONS NMS attenuates the capacity of adult hippocampal neural precursor cells to differentiate into neurons by decreasing expression of RARα through DNMT1-mediated methylation of its promoter.
Collapse
Affiliation(s)
- Shuken Boku
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York; Department of Psychiatry, Hokkaido University School of Medicine, Sapporo.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Muñiz-Hernández S, Hernández-Pedro N, Macedo-Pérez OE, Arrieta O. Alterations in Retinoic Acid Receptors in Non-Small Cell Lung Cancer and Their Clinical Implications. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/jct.2015.68072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Unlocking the potential of retinoic acid in anticancer therapy. Br J Cancer 2014; 111:2039-45. [PMID: 25412233 PMCID: PMC4260020 DOI: 10.1038/bjc.2014.412] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/03/2014] [Accepted: 06/25/2014] [Indexed: 12/20/2022] Open
Abstract
All-trans-retinoic acid (ATRA) is a physiologically active metabolite of vitamin A. Its antitumour activities have been extensively studied in a variety of model systems and clinical trials; however, to date the only malignancy responsive to ATRA treatment is acute promyelocytic leukaemia (APL) where it induces complete remission in the majority of cases when administered in combination with light chemotherapy and/or arsenic trioxide. After decades of studies, the efficacy of ATRA to treat other acute myeloid leukaemia (AML) subtypes and solid tumours remains poor. Recent studies directed to improve ATRA responsiveness in non-APL AML seem to indicate that the lack of effective ATRA response in these tumours may be primarily due to aberrant epigenetics, which negatively affect ATRA-regulated gene expression and its antileukaemic activity. Epigenetic reprogramming could potentially restore therapeutic effects of ATRA in all AML subtypes. This review discusses the current progresses in the understanding how ATRA can be utilised in the therapy of non-APL AML and other cancers.
Collapse
|
49
|
Coleman DJ, Chagani S, Hyter S, Sherman AM, Löhr CV, Liang X, Ganguli-Indra G, Indra AK. Loss of keratinocytic RXRα combined with activated CDK4 or oncogenic NRAS generates UVB-induced melanomas via loss of p53 and PTEN in the tumor microenvironment. Mol Cancer Res 2014; 13:186-96. [PMID: 25189354 DOI: 10.1158/1541-7786.mcr-14-0164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
UNLABELLED Understanding the molecular mechanisms behind formation of melanoma, the deadliest form of skin cancer, is crucial for improved diagnosis and treatment. One key is to better understand the cross-talk between epidermal keratinocytes and pigment-producing melanocytes. Here, using a bigenic mouse model system combining mutant oncogenic NRAS(Q61K) (constitutively active RAS) or mutant activated CDK4(R24C/R24C) (prevents binding of CDK4 by kinase inhibitor p16(INK4A)) with an epidermis-specific knockout of the nuclear retinoid X receptor alpha (RXRα(ep-/-)) results in increased melanoma formation after chronic ultraviolet-B (UVB) irradiation compared with control mice with functional RXRα. Melanomas from both groups of bigenic RXRα(ep-/-) mice are larger in size with higher proliferative capacity, and exhibit enhanced angiogenic properties and increased expression of malignant melanoma markers. Analysis of tumor adjacent normal skin from these mice revealed altered expression of several biomarkers indicative of enhanced melanoma susceptibility, including reduced expression of tumor suppressor p53 and loss of PTEN, with concomitant increase in activated AKT. Loss of epidermal RXRα in combination with UVB significantly enhances invasion of melanocytic cells to draining lymph nodes in bigenic mice expressing oncogenic NRAS(Q61K) compared with controls with functional RXRα. These results suggest a crucial role of keratinocytic RXRα to suppress formation of UVB-induced melanomas and their progression to malignant cancers in the context of driver mutations such as activated CDK4(R24C/R24C) or oncogenic NRAS(Q61K). IMPLICATIONS These findings suggest that RXRα may serve as a clinical diagnostic marker and therapeutic target in melanoma progression and metastasis.
Collapse
Affiliation(s)
- Daniel J Coleman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Sharmeen Chagani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Stephen Hyter
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Anna M Sherman
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. BioResource Research Program, College of Agricultural Sciences, Oregon State University, Corvallis, Oregon
| | - Christiane V Löhr
- College of Veterinary Medicine, Oregon State University, Corvallis, Oregon
| | - Xiaobo Liang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon
| | - Gitali Ganguli-Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon
| | - Arup K Indra
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, Oregon. Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon. Environmental Health Science Center, Oregon State University, Corvallis, Oregon. Department of Dermatology, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
50
|
Pan J, Guleria RS, Zhu S, Baker KM. Molecular Mechanisms of Retinoid Receptors in Diabetes-Induced Cardiac Remodeling. J Clin Med 2014; 3:566-94. [PMID: 26237391 PMCID: PMC4449696 DOI: 10.3390/jcm3020566] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 03/17/2014] [Accepted: 03/25/2014] [Indexed: 02/07/2023] Open
Abstract
Diabetic cardiomyopathy (DCM), a significant contributor to morbidity and mortality in diabetic patients, is characterized by ventricular dysfunction, in the absence of coronary atherosclerosis and hypertension. There is no specific therapeutic strategy to effectively treat patients with DCM, due to a lack of a mechanistic understanding of the disease process. Retinoic acid, the active metabolite of vitamin A, is involved in a wide range of biological processes, through binding and activation of nuclear receptors: retinoic acid receptors (RAR) and retinoid X receptors (RXR). RAR/RXR-mediated signaling has been implicated in the regulation of glucose and lipid metabolism. Recently, it has been reported that activation of RAR/RXR has an important role in preventing the development of diabetic cardiomyopathy, through improving cardiac insulin resistance, inhibition of intracellular oxidative stress, NF-κB-mediated inflammatory responses and the renin-angiotensin system. Moreover, downregulated RAR/RXR signaling has been demonstrated in diabetic myocardium, suggesting that impaired RAR/RXR signaling may be a trigger to accelerate diabetes-induced development of DCM. Understanding the molecular mechanisms of retinoid receptors in the regulation of cardiac metabolism and remodeling under diabetic conditions is important in providing the impetus for generating novel therapeutic approaches for the prevention and treatment of diabetes-induced cardiac complications and heart failure.
Collapse
Affiliation(s)
- Jing Pan
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| | - Rakeshwar S Guleria
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| | - Sen Zhu
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| | - Kenneth M Baker
- Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A & M Health Science Center, Baylor Scott & White Health, Central Texas Veterans Health Care System, Temple, TX, 76504, USA.
| |
Collapse
|