1
|
Doms S, Verlinden L, Janssens I, Vanhevel J, Eerlings R, Houtman R, Kato S, Mathieu C, Decallonne B, Carmeliet G, Verstuyf A. Coactivator-independent vitamin D receptor signaling causes severe rickets in mice, that is not prevented by a diet high in calcium, phosphate, and lactose. Bone Res 2024; 12:44. [PMID: 39164247 PMCID: PMC11335873 DOI: 10.1038/s41413-024-00343-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/26/2024] [Accepted: 05/12/2024] [Indexed: 08/22/2024] Open
Abstract
The vitamin D receptor (VDR) plays a critical role in the regulation of mineral and bone homeostasis. Upon binding of 1α,25-dihydroxyvitamin D3 to the VDR, the activation function 2 (AF2) domain repositions and recruits coactivators for the assembly of the transcriptional machinery required for gene transcription. In contrast to coactivator-induced transcriptional activation, the functional effects of coactivator-independent VDR signaling remain unclear. In humans, mutations in the AF2 domain are associated with hereditary vitamin D-resistant rickets, a genetic disorder characterized by impaired bone mineralization and growth. In the present study, we used mice with a systemic or conditional deletion of the VDR-AF2 domain (VdrΔAF2) to study coactivator-independent VDR signaling. We confirm that ligand-induced transcriptional activation was disabled because the mutant VDRΔAF2 protein was unable to interact with coactivators. Systemic VdrΔAF2 mice developed short, undermineralized bones with dysmorphic growth plates, a bone phenotype that was more pronounced than that of systemic Vdr knockout (Vdr-/-) mice. Interestingly, a rescue diet that is high in calcium, phosphate, and lactose, normalized this phenotype in Vdr-/-, but not in VdrΔAF2 mice. However, osteoblast- and osteoclast-specific VdrΔAF2 mice did not recapitulate this bone phenotype indicating coactivator-independent VDR effects are more important in other organs. In addition, RNA-sequencing analysis of duodenum and kidney revealed a decreased expression of VDR target genes in systemic VdrΔAF2 mice, which was not observed in Vdr-/- mice. These genes could provide new insights in the compensatory (re)absorption of minerals that are crucial for bone homeostasis. In summary, coactivator-independent VDR effects contribute to mineral and bone homeostasis.
Collapse
Affiliation(s)
- Stefanie Doms
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Lieve Verlinden
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Iris Janssens
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Justine Vanhevel
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Roy Eerlings
- Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
- Institute of Applied Microbiology, RWTH Aachen University, Aachen, Germany
| | | | - Shigeaki Kato
- Health Sciences Research Center, Iryo Sosei University, Iwaki, Fukuchima, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Iwaki, Fukuchima, Japan
| | - Chantal Mathieu
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Brigitte Decallonne
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Annemieke Verstuyf
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Ghosh Dastidar R, Jaroslawska J, Malinen M, Tuomainen TP, Virtanen JK, Bendik I, Carlberg C. In vivo vitamin D targets reveal the upregulation of focal adhesion-related genes in primary immune cells of healthy individuals. Sci Rep 2024; 14:17552. [PMID: 39080417 PMCID: PMC11289414 DOI: 10.1038/s41598-024-68741-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Vitamin D modulates innate and adaptive immunity, the molecular mechanisms of which we aim to understand under human in vivo conditions. Therefore, we designed the study VitDHiD (NCT03537027) as a human investigation, in which 25 healthy individuals were supplemented with a single vitamin D3 bolus (80,000 IU). Transcriptome-wide differential gene expression analysis of peripheral blood mononuclear cells (PBMCs), which were isolated directly before and 24 h after supplementation, identified 452 genes significantly (FDR < 0.05) responding to vitamin D. In vitro studies using PBMCs from the same individuals confirmed 138 of these genes as targets of 1α,25-dihydroxyvitamin D3. A subset of the 91 most regulated in vivo vitamin D target genes indicated focal adhesion as the major pathway being upregulated by vitamin D3 supplementation of healthy individuals. Differences in the individual-specific responsiveness of in vivo vitamin D target genes in relation to the increase of the person's vitamin D status allowed a segregation of the VitDHiD participants into 9 high, 12 mid and 4 low responders. The expression profile of nearly 600 genes elucidate the difference between high and low vitamin D responders, the most prominent of which is the HLA-C (major histocompatibility complex, class I, C) gene.
Collapse
Affiliation(s)
- Ranjini Ghosh Dastidar
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. J. Tuwima 10, 10-748, Olsztyn, Poland
| | - Julia Jaroslawska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. J. Tuwima 10, 10-748, Olsztyn, Poland
| | - Marjo Malinen
- Department of Forestry and Environmental Engineering, South-Eastern Finland University of Applied Sciences, Kouvola, Finland
| | - Tomi-Pekka Tuomainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Jyrki K Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Igor Bendik
- dsm-Firmenich AG, Health, Nutrition and Care (HNC), Kaiseraugst, Switzerland
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. J. Tuwima 10, 10-748, Olsztyn, Poland.
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
3
|
Jaroslawska J, Ghosh Dastidar R, Carlberg C. In vivo vitamin D target genes interconnect key signaling pathways of innate immunity. PLoS One 2024; 19:e0306426. [PMID: 39042613 PMCID: PMC11265685 DOI: 10.1371/journal.pone.0306426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/16/2024] [Indexed: 07/25/2024] Open
Abstract
The vitamin D3 metabolite 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), its nuclear receptor VDR (vitamin D receptor) and hundreds of their target genes are not only key regulators of calcium homeostasis, but also important modulators of the immune system. Innate immune cells like monocytes use VDR for efficient differentiation and are very responsive to vitamin D. So far, most information on the gene regulatory function of vitamin D and its physiological impact had been obtained from in vitro studies using supraphysiological doses of 1,25(OH)2D3. Therefore, medical experiments like the study VitDHiD (NCT03537027), where 25 healthy individuals were supplemented once with a vitamin D3 bolus (80,000 IU), provide important insight into the response to vitamin D under in vivo conditions. In this study, we inspected 452 in vivo vitamin D target genes from peripheral blood mononuclear cells (PBMCs) detected in VitDHiD and found 61 of them involved in eight major KEGG (Kyoto Encyclopedia of Genes and Genomes) pathways of innate immunity. Under in vivo conditions in healthy individuals vitamin D either silences five pathways of innate immunity, stabilizes two and increases one, so that acute inflammation is suppressed and the release of cytokines is kept under control. A ranking of the 61 target genes by inducibility, basal expression and multiple involvements in the pathways highlighted the genes NFKBIA (NFκB inhibitor alpha), NFKBIZ, FOSL2 (FOS like 2, AP1 transcription factor subunit), JDP2 (Jun dimerization protein 2), PIK3R1 (phosphoinositide-3-kinase regulatory subunit 1), CLEC7A (C-type lectin domain containing 7A), DUSP6 (dual specificity phosphatase 6), NCF2 (neutrophil cytosolic factor 2), PLCB1 (phospholipase C beta 1), PLCG2 and TNFAIP3 (TNF alpha induced protein 3). In conclusion, vitamin D's in vivo effect on innate immunity in healthy adults is mediated by the interconnection of the pathways of neutrophil extracellular trap formation, Toll-like receptor, chemokine and phagosome signaling, NOD-like receptor, C-type lectin receptor, apoptosis and interleukin 17 through a limited set of proteins encoded by key target genes.
Collapse
Affiliation(s)
- Julia Jaroslawska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Ranjini Ghosh Dastidar
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
4
|
Yeh WZ, Gresle M, Lea R, Taylor B, Lucas RM, Ponsonby AL, Mason D, Andrew J, Campbell H, Morahan J, Sampangi S, Campagna MP, Stankovich J, Van der Walt A, Jokubaitis V, Butzkueven H. The immune cell transcriptome is modulated by vitamin D 3 supplementation in people with a first demyelinating event participating in a randomized placebo-controlled trial. Clin Immunol 2024; 262:110183. [PMID: 38479439 DOI: 10.1016/j.clim.2024.110183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
Vitamin D deficiency is a risk factor for developing multiple sclerosis. The PrevANZ trial was conducted to determine if vitamin D3 supplementation can prevent recurrent disease activity in people with a first demyelinating event. As a sub-study of this trial, we investigated the effect of supplementation on peripheral immune cell gene expression. Participants were randomized to 1000, 5000 or 10,000 international units daily of vitamin D3 or placebo. Peripheral blood was collected at baseline and 12 weeks and sent for ribonucleic acid sequencing. Datasets from 55 participants were included. Gene expression was modulated by high dose supplementation. Antigen presentation and viral response pathways were upregulated. Oxidative phosphorylation and immune signaling pathways, including tumor necrosis factor-alpha and interleukin-17 signaling, were downregulated. Overall, vitamin D3 supplementation for 12 weeks modulated the peripheral immune cell transcriptome with induction of anti-inflammatory gene expression profiles. Our results support a dose-dependent effect of vitamin D3 supplementation on immune gene expression.
Collapse
Affiliation(s)
- Wei Zhen Yeh
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia.
| | - Melissa Gresle
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Rodney Lea
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia; Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Bruce Taylor
- Royal Hobart Hospital, Department of Neurology, Hobart, Australia; University of Tasmania, Menzies Institute for Medical Research, Hobart, Australia
| | - Robyn M Lucas
- Australian National University, National Centre for Epidemiology and Population Health, Canberra, Australia
| | - Anne-Louise Ponsonby
- The Florey Institute of Neuroscience and Mental Health, Early Brain Division, Parkville, Australia; University of Melbourne, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Deborah Mason
- Christchurch Hospital, Christchurch, New Zealand; New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Julie Andrew
- Neuroscience Trials Australia, Heidelberg, Australia
| | | | | | - Sandeep Sampangi
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Maria Pia Campagna
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Jim Stankovich
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anneke Van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Health, Melbourne, Victoria, Australia; MSBase Foundation, Melbourne, Australia.
| |
Collapse
|
5
|
Kilic A, Halu A, De Marzio M, Maiorino E, Duvall MG, Bruggemann TR, Rojas Quintero JJ, Chase R, Mirzakhani H, Sungur AÖ, Koepke J, Nakano T, Peh HY, Krishnamoorthy N, Abdulnour RE, Georgopoulos K, Litonjua AA, Demay M, Renz H, Levy BD, Weiss ST. Vitamin D constrains inflammation by modulating the expression of key genes on Chr17q12-21.1. eLife 2024; 12:RP89270. [PMID: 38567749 PMCID: PMC10990493 DOI: 10.7554/elife.89270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
Abstract
Vitamin D possesses immunomodulatory functions and vitamin D deficiency has been associated with the rise in chronic inflammatory diseases, including asthma (Litonjua and Weiss, 2007). Vitamin D supplementation studies do not provide insight into the molecular genetic mechanisms of vitamin D-mediated immunoregulation. Here, we provide evidence for vitamin D regulation of two human chromosomal loci, Chr17q12-21.1 and Chr17q21.2, reliably associated with autoimmune and chronic inflammatory diseases. We demonstrate increased vitamin D receptor (Vdr) expression in mouse lung CD4+ Th2 cells, differential expression of Chr17q12-21.1 and Chr17q21.2 genes in Th2 cells based on vitamin D status and identify the IL-2/Stat5 pathway as a target of vitamin D signaling. Vitamin D deficiency caused severe lung inflammation after allergen challenge in mice that was prevented by long-term prenatal vitamin D supplementation. Mechanistically, vitamin D induced the expression of the Ikzf3-encoded protein Aiolos to suppress IL-2 signaling and ameliorate cytokine production in Th2 cells. These translational findings demonstrate mechanisms for the immune protective effect of vitamin D in allergic lung inflammation with a strong molecular genetic link to the regulation of both Chr17q12-21.1 and Chr17q21.2 genes and suggest further functional studies and interventional strategies for long-term prevention of asthma and other autoimmune disorders.
Collapse
Affiliation(s)
- Ayse Kilic
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Arda Halu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Margherita De Marzio
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Environmental Health, Harvard TH Chan School of Public HealthBostonUnited States
| | - Enrico Maiorino
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Melody G Duvall
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Thayse Regina Bruggemann
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Joselyn J Rojas Quintero
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Robert Chase
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Hooman Mirzakhani
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Ayse Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-UniversityMarburgGermany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus Liebig University GiessenGiessenGermany
| | - Janine Koepke
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus Liebig University GiessenGiessenGermany
| | - Taiji Nakano
- Department of Pediatrics, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Hong Yong Peh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Nandini Krishnamoorthy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Raja-Elie Abdulnour
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Katia Georgopoulos
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children’s Hospital at Strong, University of Rochester Medical CenterRochesterUnited States
| | - Marie Demay
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University of Marburg and German Center for Lung Research (DZL)MarburgGermany
- Department of Clinical Immunology and Allergology, Laboratory of Immunopathology Sechenov UniversityMoscowRussian Federation
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| |
Collapse
|
6
|
Yeh WZ, Lea R, Stankovich J, Sampangi S, Laverick L, Van der Walt A, Jokubaitis V, Gresle M, Butzkueven H. Transcriptomics identifies blunted immunomodulatory effects of vitamin D in people with multiple sclerosis. Sci Rep 2024; 14:1436. [PMID: 38228657 PMCID: PMC10792011 DOI: 10.1038/s41598-024-51779-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024] Open
Abstract
Vitamin D deficiency is a risk factor for developing multiple sclerosis (MS). However, the immune effects of vitamin D in people with MS are not well understood. We analyzed transcriptomic datasets generated by RNA sequencing of immune cell subsets (CD4+, CD8+ T cells, B cells, monocytes) from 33 healthy controls and 33 untreated MS cases. We utilized a traditional bioinformatic pipeline and weighted gene co-expression network analysis (WGCNA) to determine genes and pathways correlated with endogenous vitamin D. In controls, CD4+ and CD8+ T cells had 1079 and 1188 genes, respectively, whose expressions were correlated with plasma 25-hydroxyvitamin D level (P < 0.05). Functional enrichment analysis identified association with TNF-alpha and MAPK signaling. In CD4+ T cells of controls, vitamin D level was associated with expression levels of several genes proximal to multiple sclerosis risk loci (P = 0.01). Genes differentially associated with endogenous vitamin D by case-control status were enriched in TNF-alpha signaling via NF-κB. WGCNA suggested a blunted response to vitamin D in cases relative to controls. Collectively, our findings provide further evidence for the immune effects of vitamin D, and demonstrate a differential immune response to vitamin D in cases relative to controls, highlighting a possible mechanism contributing to MS pathophysiology.
Collapse
Affiliation(s)
- Wei Z Yeh
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, VIC, 3004, Australia.
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia.
| | - Rodney Lea
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Jim Stankovich
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
| | - Sandeep Sampangi
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Louise Laverick
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Anneke Van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
| | - Melissa Gresle
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, VIC, 3004, Australia
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Alfred Centre, Level 6, 99 Commercial Rd, Melbourne, VIC, 3004, Australia.
- Department of Neurology, Alfred Health, Melbourne, VIC, Australia.
| |
Collapse
|
7
|
Lucock MD. Vitomics: A novel paradigm for examining the role of vitamins in human biology. Bioessays 2023; 45:e2300127. [PMID: 37727095 DOI: 10.1002/bies.202300127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023]
Abstract
The conventional view of vitamins reflects a diverse group of small molecules that facilitate critical aspects of metabolism and prevent potentially fatal deficiency syndromes. However, vitamins also contribute to the shaping and maintenance of the human phenome over lifecycle and evolutionary timescales, enabling a degree of phenotypic plasticity that operates to allow adaptive responses that are appropriate to key periods of sensitivity (i.e., epigenetic response during prenatal development within the lifecycle or as an evolved response to environmental challenge over a great many lifecycles). Individually, vitamins are important, but their effect is often based on nutrient-nutrient (vitamin-vitamin), nutrient-gene (vitamin-gene), and gene-gene interactions, and the environmental influence of shifting geophysical cycles, as well as evolving cultural practices. These ideas will be explored within what I refer to as the "adaptive vitome (vitomics)" paradigm.
Collapse
Affiliation(s)
- Mark D Lucock
- School of Environmental & Life Sciences, University of Newcastle, Ourimbah, NSW, Australia
| |
Collapse
|
8
|
Abstract
It took several hundred million years of evolution, in order to develop the endocrine vitamin D signaling system, which is formed by a nuclear receptor, the transcription factor VDR (vitamin D receptor), its ligand, the vitamin D3 metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) and several metabolizing enzymes and transport proteins. Even within the nuclear receptor superfamily the affinity of VDR for 1,25(OH)2D3 is outstandingly high (KD = 0.1 nM). The activation of VDR by 1,25(OH)2D3 is the core mechanism of genomic signaling of vitamin D3, which results in the modulation of the epigenome at thousands of promoter and enhancer regions as well as finally in the activation or repression of hundreds of target gene transcription. In addition, rapid non-genomic actions of vitamin D are described, which are mechanistically far less understood. The main function of vitamin D is to keep the human body in homeostasis. This implies the control of calcium levels, which is essential for bone mineralization, as well as for pushing of innate immunity to react sufficiently strong to microbe infection and preventing overreactions of adaptive immunity, i.e., not to cause autoimmune diseases. This review will discuss whether genomic signaling is sufficient for explaining all physiological functions of vitamin D3.
Collapse
Affiliation(s)
- Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, PL-10748 Olsztyn, Poland; School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland.
| |
Collapse
|
9
|
Carlberg C, Mycko MP. Linking Mechanisms of Vitamin D Signaling with Multiple Sclerosis. Cells 2023; 12:2391. [PMID: 37830605 PMCID: PMC10571821 DOI: 10.3390/cells12192391] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
Environmental triggers often work via signal transduction cascades that modulate the epigenome and transcriptome of cell types involved in the disease process. Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system being characterized by a combination of recurring inflammation, demyelination and progressive loss of axons. The mechanisms of MS onset are not fully understood and genetic variants may explain only some 20% of the disease susceptibility. From the environmental factors being involved in disease development low vitamin D levels have been shown to significantly contribute to MS susceptibility. The pro-hormone vitamin D3 acts via its metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) as a high affinity ligand to the transcription factor VDR (vitamin D receptor) and is a potent modulator of the epigenome at thousands of genomic regions and the transcriptome of hundreds of genes. A major target tissue of the effects of 1,25(OH)2D3 and VDR are cells of innate and adaptive immunity, such as monocytes, dendritic cells as well as B and T cells. Vitamin D induces immunological tolerance in T cells and reduces inflammatory reactions of various types of immune cells, all of which are implicated in MS pathogenesis. The immunomodulatory effects of 1,25(OH)2D3 contribute to the prevention of MS. However, the strength of the responses to vitamin D3 supplementation is highly variegated between individuals. This review will relate mechanisms of individual's vitamin D responsiveness to MS susceptibility and discuss the prospect of vitamin D3 supplementation as a way to extinguish the autoimmunity in MS.
Collapse
Affiliation(s)
- Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-748 Olsztyn, Poland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, 70211 Kuopio, Finland
| | - Marcin P. Mycko
- Department of Neurology, Laboratory of Neuroimmunology, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland;
| |
Collapse
|
10
|
Carlberg C, Raczyk M, Zawrotna N. Vitamin D: A master example of nutrigenomics. Redox Biol 2023; 62:102695. [PMID: 37043983 PMCID: PMC10119805 DOI: 10.1016/j.redox.2023.102695] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Nutrigenomics attempts to characterize and integrate the relation between dietary molecules and gene expression on a genome-wide level. One of the biologically active nutritional compounds is vitamin D3, which activates via its metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) the nuclear receptor VDR (vitamin D receptor). Vitamin D3 can be synthesized endogenously in our skin, but since we spend long times indoors and often live at higher latitudes where for many winter months UV-B radiation is too low, it became a true vitamin. The ligand-inducible transcription factor VDR is expressed in the majority of human tissues and cell types, where it modulates the epigenome at thousands of genomic sites. In a tissue-specific fashion this results in the up- and downregulation of primary vitamin D target genes, some of which are involved in attenuating oxidative stress. Vitamin D affects a wide range of physiological functions including the control of metabolism, bone formation and immunity. In this review, we will discuss how the epigenome- and transcriptome-wide effects of 1,25(OH)2D3 and its receptor VDR serve as a master example in nutrigenomics. In this context, we will outline the basis of a mechanistic understanding for personalized nutrition with vitamin D3.
Collapse
|
11
|
Moena D, Vargas E, Montecino M. Epigenetic regulation during 1,25-dihydroxyvitamin D 3-dependent gene transcription. VITAMINS AND HORMONES 2023; 122:51-74. [PMID: 36863801 DOI: 10.1016/bs.vh.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Multiple evidence accumulated over the years, demonstrates that vitamin D-dependent physiological control in vertebrates occurs primarily through the regulation of target gene transcription. In addition, there has been an increasing appreciation of the role of the chromatin organization of the genome on the ability of the active form of vitamin D, 1,25(OH)2D3, and its specific receptor VDR to regulate gene expression. Chromatin structure in eukaryotic cells is principally modulated through epigenetic mechanisms including, but not limited to, a wide number of post-translational modifications of histone proteins and ATP-dependent chromatin remodelers, which are operative in different tissues during response to physiological cues. Hence, there is necessity to understand in depth the epigenetic control mechanisms that operate during 1,25(OH)2D3-dependent gene regulation. This chapter provides a general overview about epigenetic mechanisms functioning in mammalian cells and discusses how some of these mechanisms represent important components during transcriptional regulation of the model gene system CYP24A1 in response to 1,25(OH)2D3.
Collapse
Affiliation(s)
- Daniel Moena
- School of Bachelor in Science, Faculty of Life Sciences, Universidad Andres Bello, Concepcion, Chile
| | - Esther Vargas
- School of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Martin Montecino
- Institute of Biomedical Sciences, Faculty of Medicine, Universidad Andres Bello, Santiago, Chile; Millenium Institute Center for Genome Regulation (CRG), Santiago, Chile.
| |
Collapse
|
12
|
Lucock MD. The evolution of human skin pigmentation: A changing medley of vitamins, genetic variability, and UV radiation during human expansion. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2023; 180:252-271. [PMID: 36790744 PMCID: PMC10083917 DOI: 10.1002/ajpa.24564] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 04/12/2023]
Abstract
This review examines putative, yet likely critical evolutionary pressures contributing to human skin pigmentation and subsequently, depigmentation phenotypes. To achieve this, it provides a synthesis of ideas that frame contemporary thinking, without limiting the narrative to pigmentation genes alone. It examines how geography and hence the quality and quantity of UV exposure, pigmentation genes, diet-related genes, vitamins, anti-oxidant nutrients, and cultural practices intersect and interact to facilitate the evolution of human skin color. The article has a strong focus on the vitamin D-folate evolutionary model, with updates on the latest biophysical research findings to support this paradigm. This model is examined within a broad canvas that takes human expansion out of Africa and genetic architecture into account. A thorough discourse on the biology of melanization is provided (includes relationship to BH4 and DNA damage repair), with the relevance of this to the UV sensitivity of folate and UV photosynthesis of vitamin D explained in detail, including the relevance of these vitamins to reproductive success. It explores whether we might be able to predict vitamin-related gene polymorphisms that pivot metabolism to the prevailing UVR exposome within the vitamin D-folate evolutionary hypothesis context. This is discussed in terms of a primary adaptive phenotype (pigmentation/depigmentation), a secondary adaptive phenotype (flexible metabolic phenotype based on vitamin-related gene polymorphism profile), and a tertiary adaptive strategy (dietary anti-oxidants to support the secondary adaptive phenotype). Finally, alternative evolutionary models for pigmentation are discussed, as are challenges to future research in this area.
Collapse
Affiliation(s)
- Mark D. Lucock
- School of Environmental & Life SciencesUniversity of NewcastleOurimbahNew South WalesAustralia
| |
Collapse
|
13
|
1,25(OH) 2D 3 Promotes Macrophage Efferocytosis Partly by Upregulating ASAP2 Transcription via the VDR-Bound Enhancer Region and ASAP2 May Affect Antiviral Immunity. Nutrients 2022; 14:nu14224935. [PMID: 36432619 PMCID: PMC9699620 DOI: 10.3390/nu14224935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The active form of vitamin D3, i.e., 1,25(OH)2D3, exerts an anti-inflammatory effect on the immune system, especially macrophage-mediated innate immunity. In a previous study, we identified 1,25(OH)2D3-responsive and vitamin D receptor (VDR)-bound super-enhancer regions in THP-1 cells. Herein, we examined the transcriptional regulation of ArfGAP with SH3 Domain, Ankyrin Repeat and PH Domain 2 (ASAP2) (encoding a GTPase-activating protein) by 1,25(OH)2D3 through the top-ranked VDR-bound super-enhancer region in the first intron of ASAP2 and potential functions of ASAP2 in macrophages. First, we validated the upregulation of ASAP2 by 1,25(OH)2D3 in both THP-1 cells and macrophages. Subsequently, we identified three regulatory regions (i.e., the core, 1,25(OH)2D3-responsive, and inhibitory regions) in the VDR bound-enhancer of ASAP2. ASAP2 promoted RAC1-activity and macrophage efferocytosis in vitro. Next, we assessed the functions of ASAP2 by mass spectrometry and RNA sequencing analyses. ASAP2 upregulated the expressions of antiviral-associated genes and interacted with SAM and HD domain-containing deoxynucleoside triphosphate triphosphohydrolase 1 (SAMHD1). In vivo, vitamin D reduced the number of apoptotic cells in experimental autoimmune encephalomyelitis (EAE) and promoted macrophage efferocytosis in peritonitis without changing the mRNA level of ASAP2. Thus, we could better understand the regulatory mechanism underlying ASAP2 transcription and the function of ASAP2, which may serve as a potential treatment target against inflammatory diseases and virus infections.
Collapse
|
14
|
Fernandez GJ, Ramírez-Mejia JM, Urcuqui-Inchima S. Vitamin D boosts immune response of macrophages through a regulatory network of microRNAs and mRNAs. J Nutr Biochem 2022; 109:109105. [PMID: 35858666 DOI: 10.1016/j.jnutbio.2022.109105] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 10/31/2022]
Abstract
Vitamin D is associated with the stimulation of innate immunity, inflammation, and host defense against pathogens. Macrophages express receptors of Vitamin D, regulating transcription of genes related to immune processes. However, the transcriptional and post-transcriptional strategies controlling gene expression in differentiated macrophages, and how they are influenced by Vitamin D are not well understood. We studied whether Vitamin D enhances immune response by regulating the expression of microRNAs and mRNAs. Analysis of the transcriptome showed differences in expression of 199 genes, of which 68% were up-regulated, revealing the cell state of monocyte-derived macrophages differentiated with Vitamin D (D3-MDMs) as compared to monocyte-derived macrophages (MDMs). The differentially expressed genes appear to be associated with pathophysiological processes, including inflammatory responses, and cellular stress. Transcriptional motifs in promoter regions of up- or down-regulated genes showed enrichment of VDR motifs, suggesting possible roles of transcriptional activator or repressor in gene expression. Further, microRNA-Seq analysis indicated that there were 17 differentially expressed miRNAs, of which, 7 were up-regulated and 10 down-regulated, suggesting that Vitamin D plays a critical role in the regulation of miRNA expression during macrophages differentiation. The miR-6501-3p, miR-1273h-5p, miR-665, miR-1972, miR-1183, miR-619-5p were down-regulated in D3-MDMs compared to MDMs. The integrative analysis of miRNA and mRNA expression profiles predict that miR-1972, miR-1273h-5p, and miR-665 regulate genes PDCD1LG2, IL-1B, and CD274, which are related to the inflammatory response. Results suggest an essential role of Vitamin D in macrophage differentiation that modulates host response against pathogens, inflammation, and cellular stress.
Collapse
Affiliation(s)
- Geysson Javier Fernandez
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia
| | - Julieta M Ramírez-Mejia
- Research group CIBIOP, Department of Biological Sciences, Universidad EAFIT, Medellín, Antioquia, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No 52-21, Medellín, Colombia.
| |
Collapse
|
15
|
Kashyap J, Tyagi RK. Mitotic genome bookmarking by nuclear receptor VDR advocates transmission of cellular transcriptional memory to progeny cells. Exp Cell Res 2022; 417:113193. [PMID: 35523304 DOI: 10.1016/j.yexcr.2022.113193] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 11/04/2022]
Abstract
Mitosis is an essential process for the self-renewal of cells that is accompanied by dynamic changes in nuclear architecture and chromatin organization. Despite all the changes, the cell manages to re-establish all the parental epigenetic marks, post-mitotically. Recent reports suggest that some sequence-specific transcription factors remain attached to mitotic chromatin during cell division to ensure timely reactivation of a subset of transcription factors necessary to maintain cell identity. These mitotically associated factors are suggested to act as 'genome bookmarking factors' and the phenomenon is termed 'genome bookmarking'. Here, we studied this phenomenon with Vitamin D Receptor (VDR), a key regulator of calcium and phosphate homeostasis and a member of the nuclear receptor superfamily. This study, for the first time, has confirmed VDR as a mitotic bookmarking factor that may be playing a crucial role in the maintenance of cell identity and genome bookmarking. Full 'DNA binding domain (DBD)' present in VDR was identified as essential for enrichment of VDR on mitotic chromatin. Furthermore, the study also demonstrates that VDR evokes mitotic chromatin binding behaviour in its heterodimeric partner Retinoid X receptor (RXR). Interestingly, for promoting bookmarking behaviour in RXR, both DBD and/or ligand-binding domain (LBD) in conjunction with hinge region of VDR were required. Additionally, ChIP analysis showed that VDR remains associated with DR3 (direct repeat 3) region of its specific target gene promoter CYP24A1(Cytochrome P450 family 24 subfamily A member1), during mitosis. Altogether, our study illustrates a novel function of VDR in the epigenetic transmission and control of expression of target proteome for maintenance of cell identity and traits in progeny cells.
Collapse
Affiliation(s)
- Jyoti Kashyap
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Rakesh K Tyagi
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
16
|
Warwick T, Schulz MH, Gilsbach R, Brandes RP, Seuter S. Nuclear receptor activation shapes spatial genome organization essential for gene expression control: lessons learned from the vitamin D receptor. Nucleic Acids Res 2022; 50:3745-3763. [PMID: 35325193 PMCID: PMC9023275 DOI: 10.1093/nar/gkac178] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/23/2022] [Accepted: 03/07/2022] [Indexed: 12/02/2022] Open
Abstract
Spatial genome organization is tightly controlled by several regulatory mechanisms and is essential for gene expression control. Nuclear receptors are ligand-activated transcription factors that modulate physiological and pathophysiological processes and are primary pharmacological targets. DNA binding of the important loop-forming insulator protein CCCTC-binding factor (CTCF) was modulated by 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3). We performed CTCF HiChIP assays to produce the first genome-wide dataset of CTCF long-range interactions in 1,25(OH)2D3-treated cells, and to determine whether dynamic changes of spatial chromatin interactions are essential for fine-tuning of nuclear receptor signaling. We detected changes in 3D chromatin organization upon vitamin D receptor (VDR) activation at 3.1% of all observed CTCF interactions. VDR binding was enriched at both differential loop anchors and within differential loops. Differential loops were observed in several putative functional roles including TAD border formation, promoter-enhancer looping, and establishment of VDR-responsive insulated neighborhoods. Vitamin D target genes were enriched in differential loops and at their anchors. Secondary vitamin D effects related to dynamic chromatin domain changes were linked to location of downstream transcription factors in differential loops. CRISPR interference and loop anchor deletion experiments confirmed the functional relevance of nuclear receptor ligand-induced adjustments of the chromatin 3D structure for gene expression regulation.
Collapse
Affiliation(s)
- Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt/Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main 60590, Frankfurt am Main, Germany
| | - Marcel H Schulz
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt/Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main 60590, Frankfurt am Main, Germany
| | - Ralf Gilsbach
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt/Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main 60590, Frankfurt am Main, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt/Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main 60590, Frankfurt am Main, Germany
| | - Sabine Seuter
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt/Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main 60590, Frankfurt am Main, Germany
| |
Collapse
|
17
|
Vitamin D and Its Target Genes. Nutrients 2022; 14:nu14071354. [PMID: 35405966 PMCID: PMC9003440 DOI: 10.3390/nu14071354] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/22/2022] Open
Abstract
The vitamin D metabolite 1α,25-dihydroxyvitamin D3 is the natural, high-affinity ligand of the transcription factor vitamin D receptor (VDR). In many tissues and cell types, VDR binds in a ligand-dependent fashion to thousands of genomic loci and modulates, via local chromatin changes, the expression of hundreds of primary target genes. Thus, the epigenome and transcriptome of VDR-expressing cells is directly affected by vitamin D. Vitamin D target genes encode for proteins with a large variety of physiological functions, ranging from the control of calcium homeostasis, innate and adaptive immunity, to cellular differentiation. This review will discuss VDR’s binding to genomic DNA, as well as its genome-wide locations and interaction with partner proteins, in the context of chromatin. This information will be integrated into a model of vitamin D signaling, explaining the regulation of vitamin D target genes.
Collapse
|
18
|
Time-Resolved Gene Expression Analysis Monitors the Regulation of Inflammatory Mediators and Attenuation of Adaptive Immune Response by Vitamin D. Int J Mol Sci 2022; 23:ijms23020911. [PMID: 35055093 PMCID: PMC8776203 DOI: 10.3390/ijms23020911] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/13/2022] [Indexed: 02/08/2023] Open
Abstract
Peripheral blood mononuclear cells (PBMCs) belong to the innate and adaptive immune system and are highly sensitive and responsive to changes in their systemic environment. In this study, we focused on the time course of transcriptional changes in freshly isolated human PBMCs 4, 8, 24 and 48 h after onset of stimulation with the active vitamin D metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3). Taking all four time points together, 662 target genes were identified and segregated either by time of differential gene expression into 179 primary and 483 secondary targets or by driver of expression change into 293 direct and 369 indirect targets. The latter classification revealed that more than 50% of target genes were primarily driven by the cells' response to ex vivo exposure than by the nuclear hormone and largely explained its down-regulatory effect. Functional analysis indicated vitamin D's role in the suppression of the inflammatory and adaptive immune response by down-regulating ten major histocompatibility complex class II genes, five alarmins of the S100 calcium binding protein A family and by affecting six chemokines of the C-X-C motif ligand family. Taken together, studying time-resolved responses allows to better contextualize the effects of vitamin D on the immune system.
Collapse
|
19
|
Bikle DD. Ligand-Independent Actions of the Vitamin D Receptor: More Questions Than Answers. JBMR Plus 2021; 5:e10578. [PMID: 34950833 PMCID: PMC8674770 DOI: 10.1002/jbm4.10578] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/26/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Our predominant understanding of the actions of vitamin D involve binding of its ligand, 1,25(OH)D, to the vitamin D receptor (VDR), which for its genomic actions binds to discrete regions of its target genes called vitamin D response elements. However, chromatin immunoprecipitation‐sequencing (ChIP‐seq) studies have observed that the VDR can bind to many sites in the genome without its ligand. The number of such sites and how much they coincide with sites that also bind the liganded VDR vary from cell to cell, with the keratinocyte from the skin having the greatest overlap and the intestinal epithelial cell having the least. What is the purpose of the unliganded VDR? In this review, I will focus on two clear examples in which the unliganded VDR plays a role. The best example is that of hair follicle cycling. Hair follicle cycling does not need 1,25(OH)2D, and Vdr lacking the ability to bind 1,25(OH)2D can restore hair follicle cycling in mice otherwise lacking Vdr. This is not true for other functions of VDR such as intestinal calcium transport. Tumor formation in the skin after UVB radiation or the application of chemical carcinogens also appears to be at least partially independent of 1,25(OH)2D in that Vdr null mice develop such tumors after these challenges, but mice lacking Cyp27b1, the enzyme producing 1,25(OH)2D, do not. Examples in other tissues emerge when studies comparing Vdr null and Cyp27b1 null mice are compared, demonstrating a more severe phenotype with respect to bone mineral homeostasis in the Cyp27b1 null mouse, suggesting a repressor function for VDR. This review will examine potential mechanisms for these ligand‐independent actions of VDR, but as the title indicates, there are more questions than answers with respect to this role of VDR. © 2021 The Author. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Daniel D Bikle
- Departments of Medicine and Dermatology University of California San Francisco, San Francisco VA Health Center San Francisco CA USA
| |
Collapse
|
20
|
Sabatier M, Boet E, Zaghdoudi S, Guiraud N, Hucteau A, Polley N, Cognet G, Saland E, Lauture L, Farge T, Sahal A, Pancaldi V, Chu-Van E, Castelli F, Bertoli S, Bories P, Récher C, Boutzen H, Mansat-De Mas V, Stuani L, Sarry JE. Activation of Vitamin D Receptor Pathway Enhances Differentiating Capacity in Acute Myeloid Leukemia with Isocitrate Dehydrogenase Mutations. Cancers (Basel) 2021; 13:5243. [PMID: 34680392 PMCID: PMC8533831 DOI: 10.3390/cancers13205243] [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: 08/25/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 11/30/2022] Open
Abstract
Relapses and resistance to therapeutic agents are major barriers in the treatment of acute myeloid leukemia (AML) patients. These unfavorable outcomes emphasize the need for new strategies targeting drug-resistant cells. As IDH mutations are present in the preleukemic stem cells and systematically conserved at relapse, targeting IDH mutant cells could be essential to achieve a long-term remission in the IDH mutant AML subgroup. Here, using a panel of human AML cell lines and primary AML patient specimens harboring IDH mutations, we showed that the production of an oncometabolite (R)-2-HG by IDH mutant enzymes induces vitamin D receptor-related transcriptional changes, priming these AML cells to differentiate with pharmacological doses of ATRA and/or VD. This activation occurs in a CEBPα-dependent manner. Accordingly, our findings illuminate potent and cooperative effects of IDH mutations and the vitamin D receptor pathway on differentiation in AML, revealing a novel therapeutic approach easily transferable/immediately applicable to this subgroup of AML patients.
Collapse
Affiliation(s)
- Marie Sabatier
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Emeline Boet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Sonia Zaghdoudi
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Nathan Guiraud
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Alexis Hucteau
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Nathaniel Polley
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Guillaume Cognet
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Estelle Saland
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Laura Lauture
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Thomas Farge
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Ambrine Sahal
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Vera Pancaldi
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
| | - Emeline Chu-Van
- CEA/DSV/iBiTec-S/SPI, Laboratoire d’Etude du Métabolisme des Médicaments, MetaboHUB-Paris, 91191 Gif-sur-Yvette, France; (E.C.-V.); (F.C.)
| | - Florence Castelli
- CEA/DSV/iBiTec-S/SPI, Laboratoire d’Etude du Métabolisme des Médicaments, MetaboHUB-Paris, 91191 Gif-sur-Yvette, France; (E.C.-V.); (F.C.)
| | - Sarah Bertoli
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
- Département d’Hématologie, University of Toulouse, CEDEX 6, 31013 Toulouse, France
- Service d’Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, CEDEX 9, 31059 Toulouse, France
| | - Pierre Bories
- Réseau Régional de Cancérologie Onco-Occitanie, CEDEX 9, 31059 Toulouse, France;
| | - Christian Récher
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
- Département d’Hématologie, University of Toulouse, CEDEX 6, 31013 Toulouse, France
- Service d’Hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, CHU de Toulouse, CEDEX 9, 31059 Toulouse, France
| | - Héléna Boutzen
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Véronique Mansat-De Mas
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
- Département d’Hématologie, University of Toulouse, CEDEX 6, 31013 Toulouse, France
| | - Lucille Stuani
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| | - Jean-Emmanuel Sarry
- Centre de Recherches en Cancérologie de Toulouse, Université de Toulouse, Inserm, Centre National de Recherche Scientifique, CEDEX 1, 31037 Toulouse, France; (M.S.); (E.B.); (S.Z.); (N.G.); (A.H.); (N.P.); (G.C.); (E.S.); (L.L.); (T.F.); (A.S.); (V.P.); (S.B.); (C.R.); (H.B.); (V.M.-D.M.)
- LabEx Toucan, 31037 Toulouse, France
- Equipe Labellisée Ligue Nationale Contre le Cancer 2018, 31037 Toulouse, France
| |
Collapse
|
21
|
Kalia V, Studzinski GP, Sarkar S. Role of vitamin D in regulating COVID-19 severity-An immunological perspective. J Leukoc Biol 2021; 110:809-819. [PMID: 33464639 PMCID: PMC8014852 DOI: 10.1002/jlb.4covr1020-698r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/30/2020] [Accepted: 12/30/2020] [Indexed: 12/23/2022] Open
Abstract
Vitamin D, a key nutrient/prohormone classically associated with skeletal health, is also an important immunomodulator, with pleotropic effects on innate and adaptive immune cells. Outcomes of several chronic, autoimmune, and infectious diseases are linked to vitamin D. Emergent correlations of vitamin D insufficiency with coronavirus-induced disease 2019 (COVID-19) severity, alongside empirical and clinical evidence of immunoregulation by vitamin D in other pulmonary diseases, have prompted proposals of vitamin D supplementation to curb the COVID-19 public health toll. In this review paper, we engage an immunological lens to discuss potential mechanisms by which vitamin D signals might regulate respiratory disease severity in severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infections, vis a vis other pulmonary infections. It is proposed that vitamin D signals temper lung inflammatory cascades during SARS-CoV2 infection, and insufficiency of vitamin D causes increased inflammatory cytokine storm, thus leading to exacerbated respiratory disease. Additionally, analogous to studies of reduced cancer incidence, the dosage of vitamin D compounds administered to patients near the upper limit of safety may serve to maximize immune health benefits and mitigate inflammation and disease severity in SARS-CoV2 infections. We further deliberate on the importance of statistically powered clinical correlative and interventional studies, and the need for in-depth basic research into vitamin D-dependent host determinants of respiratory disease severity.
Collapse
Affiliation(s)
- Vandana Kalia
- Department of Pediatrics, Division of Hematology and Oncology, University of Washington School of Medicine, Seattle, Washington, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - George P Studzinski
- Department of Pathology, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Surojit Sarkar
- Department of Pediatrics, Division of Hematology and Oncology, University of Washington School of Medicine, Seattle, Washington, USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
22
|
Vitamin D and the risk for cancer: A molecular analysis. Biochem Pharmacol 2021; 196:114735. [PMID: 34411566 DOI: 10.1016/j.bcp.2021.114735] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 02/08/2023]
Abstract
Uncontrolled overgrowth of cells, such as in cancer, is an unavoidable risk in life that affects nearly every second individual in industrialized countries. However, in part this risk can be controlled through lifestyle adjustments, such as the avoidance of smoking, unhealthy diet, obesity, physical inactivity and other cancer risk factors. A low vitaminD status is a risk in particular for cancers of colon, prostate, breast and leukocytes. VitaminD3 is produced non-enzymatically, when the cholesterol precursor 7-dehydrocholesterol is exposed to UV-B from sunlight, i.e., all cholesterol synthesizing species, including humans, can make vitaminD3. VitaminD endocrinology started some 550million years ago, when the metabolite 1α,25-dihydroxyvitaminD3 and the transcription factor vitaminD receptor teamed up for regulating the expression of hundreds of target genes in a multitude of different tissues and cell types. Initially, these genes were focused on the control of energy homeostasis, which later also involved energy-demanding innate and adaptive immunity. Rapidly growing cells of the immune system as well as those of malignant tumors rely on comparable genes and pathways, some of which are modulated by vitaminD. Accordingly, vitaminD has anti-cancer effects both directly via controling the differentiation, proliferation and apoptosis of neoplastic cells as well as indirectly through regulating immune cells that belong to the microenvironment of malignant tumors. This review discusses effects of vitaminD on the epigenome and transcriptome of stromal and tumor cells, inter-individual variations in vitaminD responsiveness and their relation to the prevention and possible therapy of cancer.
Collapse
|
23
|
A hierarchical regulatory network analysis of the vitamin D induced transcriptome reveals novel regulators and complete VDR dependency in monocytes. Sci Rep 2021; 11:6518. [PMID: 33753848 PMCID: PMC7985518 DOI: 10.1038/s41598-021-86032-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/04/2021] [Indexed: 11/11/2022] Open
Abstract
The transcription factor vitamin D receptor (VDR) is the high affinity nuclear target of the biologically active form of vitamin D3 (1,25(OH)2D3). In order to identify pure genomic transcriptional effects of 1,25(OH)2D3, we used VDR cistrome, transcriptome and open chromatin data, obtained from the human monocytic cell line THP-1, for a novel hierarchical analysis applying three bioinformatics approaches. We predicted 75.6% of all early 1,25(OH)2D3-responding (2.5 or 4 h) and 57.4% of the late differentially expressed genes (24 h) to be primary VDR target genes. VDR knockout led to a complete loss of 1,25(OH)2D3–induced genome-wide gene regulation. Thus, there was no indication of any VDR-independent non-genomic actions of 1,25(OH)2D3 modulating its transcriptional response. Among the predicted primary VDR target genes, 47 were coding for transcription factors and thus may mediate secondary 1,25(OH)2D3 responses. CEBPA and ETS1 ChIP-seq data and RNA-seq following CEBPA knockdown were used to validate the predicted regulation of secondary vitamin D target genes by both transcription factors. In conclusion, a directional network containing 47 partly novel primary VDR target transcription factors describes secondary responses in a highly complex vitamin D signaling cascade. The central transcription factor VDR is indispensable for all transcriptome-wide effects of the nuclear hormone.
Collapse
|
24
|
Lucock M. Vitamin-related phenotypic adaptation to exposomal factors: The folate-vitamin D-exposome triad. Mol Aspects Med 2021; 87:100944. [PMID: 33551238 DOI: 10.1016/j.mam.2021.100944] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/02/2021] [Accepted: 01/25/2021] [Indexed: 12/16/2022]
Abstract
The biological role of two key vitamins, folic acid and vitamin D is so fundamental to life processes, it follows that their UV sensitivity, dietary abundance (both key exposomal factors) and variability in dependent genes will modify their functional efficacy, particularly in the context of maintaining the integrity and function of genome and epigenome. This article therefore examines folate and vitamin D-related phenotypic adaptation to environmental factors which vary across the human life cycle as well as over an evolutionary time-scale. Molecular mechanisms, key nutrigenomic factors, phenotypic maladaptation and evolutionary models are discussed.
Collapse
Affiliation(s)
- Mark Lucock
- School of Environmental & Life Sciences, University of Newcastle, PO Box 127, Brush Rd, Ourimbah, NSW, 2258, Australia.
| |
Collapse
|
25
|
Common and personal target genes of the micronutrient vitamin D in primary immune cells from human peripheral blood. Sci Rep 2020; 10:21051. [PMID: 33273683 PMCID: PMC7713372 DOI: 10.1038/s41598-020-78288-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022] Open
Abstract
Vitamin D is essential for the function of the immune system. In this study, we treated peripheral blood mononuclear cells (PBMCs) of healthy adults with the biologically active form of vitamin D3, 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) using two different approaches: single repeats with PBMCs obtained from a cohort of 12 individuals and personalized analysis based on triplicates of five study participants. This identified 877 (cohort approach) and 3951 (personalized approach) genes that significantly (p < 0.05) changed their expression 24 h after 1,25(OH)2D3 stimulation. From these, 333 and 1232 were classified as supertargets, a third of which were identified as novel. Individuals differed largely in their vitamin D response not only by the magnitude of expression change but also by their personal selection of (super)target genes. Functional analysis of the target genes suggested the overarching role of vitamin D in the regulation of metabolism, proliferation and differentiation, but in particular in the control of functions mediated by the innate and adaptive immune system, such as responses to infectious diseases and chronic inflammatory disorders. In conclusion, immune cells are an important target of vitamin D and common genes may serve as biomarkers for personal responses to the micronutrient.
Collapse
|
26
|
Yeh WZ, Gresle M, Jokubaitis V, Stankovich J, van der Walt A, Butzkueven H. Immunoregulatory effects and therapeutic potential of vitamin D in multiple sclerosis. Br J Pharmacol 2020; 177:4113-4133. [PMID: 32668009 DOI: 10.1111/bph.15201] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/19/2022] Open
Abstract
Initially recognised as an important factor for bone health, vitamin D is now known to have a range of effects on the immune system. Vitamin D deficiency is associated with an increased risk of multiple sclerosis (MS), a chronic immune-mediated demyelinating disease of the CNS. In this review, we explore the links between vitamin D deficiency, MS risk, and disease activity. We also discuss the known immune effects of vitamin D supplementation and the relevance of these observations to the immunopathology of MS. Finally, we review the existing evidence for vitamin D supplementation as an MS therapy, highlighting several recent clinical studies and trials.
Collapse
Affiliation(s)
- Wei Zhen Yeh
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Melissa Gresle
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Jim Stankovich
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Anneke van der Walt
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Glinsky GV. Tripartite Combination of Candidate Pandemic Mitigation Agents: Vitamin D, Quercetin, and Estradiol Manifest Properties of Medicinal Agents for Targeted Mitigation of the COVID-19 Pandemic Defined by Genomics-Guided Tracing of SARS-CoV-2 Targets in Human Cells. Biomedicines 2020; 8:E129. [PMID: 32455629 PMCID: PMC7277789 DOI: 10.3390/biomedicines8050129] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022] Open
Abstract
Genes required for SARS-CoV-2 entry into human cells, ACE2 and FURIN, were employed as baits to build genomic-guided molecular maps of upstream regulatory elements, their expression and functions in the human body, and pathophysiologically relevant cell types. Repressors and activators of the ACE2 and FURIN genes were identified based on the analyses of gene silencing and overexpression experiments as well as relevant transgenic mouse models. Panels of repressors (VDR; GATA5; SFTPC; HIF1a) and activators (HMGA2; INSIG1; RUNX1; HNF4a; JNK1/c-FOS) were then employed to identify existing drugs manifesting in their effects on gene expression signatures of potential coronavirus infection mitigation agents. Using this strategy, vitamin D and quercetin have been identified as putative 2019 coronavirus disease (COVID-19) mitigation agents. Quercetin has been identified as one of top-scoring candidate therapeutics in the supercomputer SUMMIT drug-docking screen and Gene Set Enrichment Analyses (GSEA) of expression profiling experiments (EPEs), indicating that highly structurally similar quercetin, luteolin, and eriodictyol could serve as scaffolds for the development of efficient inhibitors of SARS-CoV-2 infection. In agreement with this notion, quercetin alters the expression of 98 of 332 (30%) of human genes encoding protein targets of SARS-CoV-2, thus potentially interfering with functions of 23 of 27 (85%) of the SARS-CoV-2 viral proteins in human cells. Similarly, Vitamin D may interfere with functions of 19 of 27 (70%) of the SARS-CoV-2 proteins by altering expression of 84 of 332 (25%) of human genes encoding protein targets of SARS-CoV-2. Considering the potential effects of both quercetin and vitamin D, the inference could be made that functions of 25 of 27 (93%) of SARS-CoV-2 proteins in human cells may be altered. GSEA and EPEs identify multiple drugs, smoking, and many disease conditions that appear to act as putative coronavirus infection-promoting agents. Discordant patterns of testosterone versus estradiol impacts on SARS-CoV-2 targets suggest a plausible molecular explanation of the apparently higher male mortality during the coronavirus pandemic. Estradiol, in contrast with testosterone, affects the expression of the majority of human genes (203 of 332; 61%) encoding SARS-CoV-2 targets, thus potentially interfering with functions of 26 of 27 SARS-CoV-2 viral proteins. A hypothetical tripartite combination consisting of quercetin/vitamin D/estradiol may affect expression of 244 of 332 (73%) human genes encoding SARS-CoV-2 targets. Of major concern is the ACE2 and FURIN expression in many human cells and tissues, including immune cells, suggesting that SARS-CoV-2 may infect a broad range of cellular targets in the human body. Infection of immune cells may cause immunosuppression, long-term persistence of the virus, and spread of the virus to secondary targets. Present analyses and numerous observational studies indicate that age-associated vitamin D deficiency may contribute to the high mortality of older adults and the elderly. Immediate availability for targeted experimental and clinical interrogations of potential COVID-19 pandemic mitigation agents, namely vitamin D and quercetin, as well as of the highly selective (Ki, 600 pm) intrinsically specific FURIN inhibitor (a1-antitrypsin Portland (a1-PDX), is considered an encouraging factor. Observations reported in this contribution are intended to facilitate follow-up targeted experimental studies and, if warranted, randomized clinical trials to identify and validate therapeutically viable interventions to combat the COVID-19 pandemic. Specifically, gene expression profiles of vitamin D and quercetin activities and their established safety records as over-the-counter medicinal substances strongly argue that they may represent viable candidates for further considerations of their potential utility as COVID-19 pandemic mitigation agents. In line with the results of present analyses, a randomized interventional clinical trial evaluating effects of estradiol on severity of the coronavirus infection in COVID19+ and presumptive COVID19+ patients and two interventional randomized clinical trials evaluating effects of vitamin D on prevention and treatment of COVID-19 were listed on the ClinicalTrials.gov website.
Collapse
Affiliation(s)
- Gennadi V Glinsky
- Institute of Engineering in Medicine, University of California, San Diego, 9500 Gilman Dr. MC 0435, La Jolla, CA 92093-0435, USA
| |
Collapse
|
28
|
Impact of Epigenetics on Complications of Fanconi Anemia: The Role of Vitamin D-Modulated Immunity. Nutrients 2020; 12:nu12051355. [PMID: 32397406 PMCID: PMC7285109 DOI: 10.3390/nu12051355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/02/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Fanconi anemia (FA) is a rare disorder with the clinical characteristics of (i) specific malformations at birth, (ii) progressive bone marrow failure already during early childhood and (iii) dramatically increased risk of developing cancer in early age, such as acute myeloid leukemia and squamous cell carcinoma. Patients with FA show DNA fragility due to a defect in the DNA repair machinery based on predominately recessive mutations in 23 genes. Interestingly, patients originating from the same family and sharing an identical mutation, frequently show significant differences in their clinical presentation. This implies that epigenetics plays an important role in the manifestation of the disease. The biologically active form of vitamin D, 1α,25-dihydroxyvitamin D3 controls cellular growth, differentiation and apoptosis via the modulation of the immune system. The nuclear hormone activates the transcription factor vitamin D receptor that affects, via fine-tuning of the epigenome, the transcription of >1000 human genes. In this review, we discuss that changes in the epigenome, in particular in immune cells, may be central for the clinical manifestation of FA. These epigenetic changes can be modulated by vitamin D suggesting that the individual FA patient’s vitamin D status and responsiveness are of critical importance for disease progression.
Collapse
|
29
|
Carlberg C, Muñoz A. An update on vitamin D signaling and cancer. Semin Cancer Biol 2020; 79:217-230. [DOI: 10.1016/j.semcancer.2020.05.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/21/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022]
|
30
|
Koivisto O, Hanel A, Carlberg C. Key Vitamin D Target Genes with Functions in the Immune System. Nutrients 2020; 12:E1140. [PMID: 32325790 PMCID: PMC7230898 DOI: 10.3390/nu12041140] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/18/2022] Open
Abstract
The biologically active form of vitamin D3, 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3), modulates innate and adaptive immunity via genes regulated by the transcription factor vitamin D receptor (VDR). In order to identify the key vitamin D target genes involved in these processes, transcriptome-wide datasets were compared, which were obtained from a human monocytic cell line (THP-1) and peripheral blood mononuclear cells (PBMCs) treated in vitro by 1,25(OH)2D3, filtered using different approaches, as well as from PBMCs of individuals supplemented with a vitamin D3 bolus. The led to the genes ACVRL1, CAMP, CD14, CD93, CEBPB, FN1, MAPK13, NINJ1, LILRB4, LRRC25, SEMA6B, SRGN, THBD, THEMIS2 and TREM1. Public epigenome- and transcriptome-wide data from THP-1 cells were used to characterize these genes based on the level of their VDR-driven enhancers as well as the level of the dynamics of their mRNA production. Both types of datasets allowed the categorization of the vitamin D target genes into three groups according to their role in (i) acute response to infection, (ii) infection in general and (iii) autoimmunity. In conclusion, 15 genes were identified as major mediators of the action of vitamin D in innate and adaptive immunity and their individual functions are explained based on different gene regulatory scenarios.
Collapse
Affiliation(s)
| | | | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (O.K.); (A.H.)
| |
Collapse
|
31
|
Carlberg C. Vitamin D: A Micronutrient Regulating Genes. Curr Pharm Des 2020; 25:1740-1746. [PMID: 31298160 DOI: 10.2174/1381612825666190705193227] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/21/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND At sufficient sun exposure, humans can synthesize vitamin D3 endogenously in their skin, but today's lifestyle makes the secosteroid a true vitamin that needs to be taken up by diet or supplementation with pills. The vitamin D3 metabolite 1α,25-dihydroxyvitamin D3 acts as a nuclear hormone activating the transcription factor vitamin D receptor (VDR). METHODS This review discusses the biological effects of micronutrient vitamin D ranging from calcium homeostasis and bone formation to the modulation of innate and adaptive immunity. RESULTS Since normal human diet is sufficient in vitamin D, the need for efficient vitamin D3 synthesis in the skin acts as an evolutionary driver for its lightening during the migration out of Africa towards North. Via activating the VDR, vitamin D has direct effects on the epigenome and the expression of more than 1000 genes in most human tissues and cell types. CONCLUSIONS The pleiotropic action of vitamin D in health and disease prevention is explained through complex gene regulatory events of the transcription factor VDR.
Collapse
Affiliation(s)
- Carsten Carlberg
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
32
|
Carlberg C. Vitamin D Signaling in the Context of Innate Immunity: Focus on Human Monocytes. Front Immunol 2019; 10:2211. [PMID: 31572402 PMCID: PMC6753645 DOI: 10.3389/fimmu.2019.02211] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022] Open
Abstract
The vitamin D3 metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) activates at sub-nanomolar concentrations the transcription factor vitamin D receptor (VDR). VDR is primarily involved in the control of cellular metabolism but in addition modulates processes important for immunity, such as anti-microbial defense and the induction of T cell tolerance. Monocytes and their differentiated phenotypes, macrophages and dendritic cells, are key cell types of the innate immune system, in which vitamin D signaling was most comprehensively investigated via the use of next generation sequencing technologies. These investigations provided genome-wide maps illustrating significant effects of 1,25(OH)2D3 on the binding of VDR, the pioneer transcription factors purine-rich box 1 (PU.1) and CCAAT/enhancer binding protein α (CEBPA) and the chromatin modifier CCCTC-binding factor (CTCF) as well as on chromatin accessibility and histone markers of promoter and enhancer regions, H3K4me3 and H3K27ac. Thus, the epigenome of human monocytes is at multiple levels sensitive to vitamin D. These data served as the basis for the chromatin model of vitamin D signaling, which mechanistically explains the activation of a few hundred primary vitamin D target genes. Comparable epigenome- and transcriptome-wide effects of vitamin D were also described in peripheral blood mononuclear cells isolated from individuals before and after supplementation with a vitamin D3 bolus. This review will conclude with the hypothesis that vitamin D modulates the epigenome of immune cells during perturbations by antigens and other immunological challenges suggesting that an optimal vitamin D status may be essential for an effective epigenetic learning process, in particular of the innate immune system.
Collapse
Affiliation(s)
- Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
33
|
Fleet JC, Kovalenko PL, Li Y, Smolinski J, Spees C, Yu JG, Thomas-Ahner JM, Cui M, Neme A, Carlberg C, Clinton SK. Vitamin D Signaling Suppresses Early Prostate Carcinogenesis in TgAPT 121 Mice. Cancer Prev Res (Phila) 2019; 12:343-356. [PMID: 31028080 DOI: 10.1158/1940-6207.capr-18-0401] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/03/2019] [Accepted: 04/22/2019] [Indexed: 12/14/2022]
Abstract
We tested whether lifelong modification of vitamin D signaling can alter the progression of early prostate carcinogenesis in studies using mice that develop high-grade prostatic intraepithelial neoplasia that is similar to humans. Two tissue-limited models showed that prostate vitamin D receptor (VDR) loss increased prostate carcinogenesis. In another study, we fed diets with three vitamin D3 levels (inadequate = 25 IU/kg diet, adequate for bone health = 150 IU/kg, or high = 1,000 IU/kg) and two calcium levels (adequate for bone health = 0.5% and high = 1.5%). Dietary vitamin D caused a dose-dependent increase in serum 25-hydroxyvitamin D levels and a reduction in the percentage of mice with adenocarcinoma but did not improve bone mass. In contrast, high calcium suppressed serum 1,25-dihydroxyvitamin D levels and improved bone mass but increased the incidence of adenocarcinoma. Analysis of the VDR cistrome in RWPE1 prostate epithelial cells revealed vitamin D-mediated regulation of multiple cancer-relevant pathways. Our data support the hypothesis that the loss of vitamin D signaling accelerates the early stages of prostate carcinogenesis, and our results suggest that different dietary requirements may be needed to support prostate health or maximize bone mass. SIGNIFICANCE: This work shows that disrupting vitamin D signaling through diet or genetic deletion increases early prostate carcinogenesis through multiple pathways. Higher-diet vitamin D levels are needed for cancer than bone.
Collapse
Affiliation(s)
- James C Fleet
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana. .,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - Pavlo L Kovalenko
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Yan Li
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Justin Smolinski
- Division of Medical Oncology, College of Medicine, Columbus, Ohio
| | - Colleen Spees
- Division of Medical Oncology, College of Medicine, Columbus, Ohio
| | - Jun-Ge Yu
- Division of Medical Oncology, College of Medicine, Columbus, Ohio
| | | | - Min Cui
- Department of Nutrition Science, Purdue University, West Lafayette, Indiana
| | - Antonio Neme
- Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas-Mérida, Universidad Nacional Autónoma de México, Yucatán, México
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Steven K Clinton
- Division of Medical Oncology, College of Medicine, Columbus, Ohio.,The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
34
|
Neme A, Seuter S, Malinen M, Nurmi T, Tuomainen TP, Virtanen JK, Carlberg C. In vivo transcriptome changes of human white blood cells in response to vitamin D. J Steroid Biochem Mol Biol 2019; 188:71-76. [PMID: 30537545 DOI: 10.1016/j.jsbmb.2018.11.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 12/31/2022]
Abstract
In the vitamin D intervention study VitDbol (NCT02063334) blood samples were drawn directly before an oral bolus (2000 μg vitamin D3) and 24 h later. The focus of phase II of VitDbol was the transcriptome-wide analysis of the effects of vitamin D gene expression in human peripheral blood mononuclear cells (PBMCs). All five participants responded in an individual fashion to the bolus by increases in serum levels of the vitamin D metabolites 25-hydroxyvitamin D3 (25(OH)D3) and 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3). RNA sequencing identified 15.040 commonly expressed genes in PBMCs, 702 (4,7%) of which were significantly (p < 0,05) affected by the vitamin D3 bolus. KEGG pathway analysis suggested that these genes are involved in general protein translation, monocyte differentiation and cellular growth control. Previously published transcriptome-wide studies in comparable cell systems confirmed 234 of the 702 vitamin D target genes, leaving many genes, such as HLA-A and HLA-C, as novel discoveries. Interestingly, in vivo stimulated PBMCs of this study showed a larger number of common vitamin D target genes with the monocytic cell line THP-1 than with in vitro stimulated PBMCs. The expression pattern of vitamin D target genes differed significantly between individuals and the average expression change can serve as a marker for vitamin D responsiveness. In conclusion, this study demonstrates that under in vivo conditions changes in 25(OH)D3 and 1,25(OH)2D3 serum concentrations alter the expression of more than 700 vitamin D target genes in human leukocytes.
Collapse
Affiliation(s)
- Antonio Neme
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Sabine Seuter
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Marjo Malinen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Tarja Nurmi
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Tomi-Pekka Tuomainen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Jyrki K Virtanen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
35
|
Abstract
Nutrigenomics studies how environmental factors, such as food intake and lifestyle, influence the expression of the genome. Vitamin D₃ represents a master example of nutrigenomics, since via its metabolite 1α,25-dihydroxyvitamin D₃, which binds with high-affinity to the vitamin D receptor, the secosteroid directly affects the epigenome and transcriptome at thousands of loci within the human genome. Vitamin D is important for both cellular metabolism and immunity, as it controls calcium homeostasis and modulates the response of the innate and adaptive immune system. At sufficient UV-B exposure, humans can synthesize vitamin D₃ endogenously in their skin, but today's lifestyle often makes the molecule a true vitamin and micronutrient that needs to be taken up by diet or supplementation with pills. The individual's molecular response to vitamin D requires personalized supplementation with vitamin D₃, in order to obtain optimized clinical benefits in the prevention of osteoporosis, sarcopenia, autoimmune diseases, and possibly different types of cancer. The importance of endogenous synthesis of vitamin D₃ created an evolutionary pressure for reduced skin pigmentation, when, during the past 50,000 years, modern humans migrated from Africa towards Asia and Europe. This review will discuss different aspects of how vitamin D interacts with the human genome, focusing on nutritional epigenomics in context of immune responses. This should lead to a better understanding of the clinical benefits of vitamin D.
Collapse
Affiliation(s)
- Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland.
| |
Collapse
|
36
|
Lu M, McComish BJ, Burdon KP, Taylor BV, Körner H. The Association Between Vitamin D and Multiple Sclerosis Risk: 1,25(OH) 2D 3 Induces Super-Enhancers Bound by VDR. Front Immunol 2019; 10:488. [PMID: 30941131 PMCID: PMC6433938 DOI: 10.3389/fimmu.2019.00488] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/22/2019] [Indexed: 11/16/2022] Open
Abstract
A super-enhancer (SE) is a cluster of enhancers with a relatively high density of particular chromatin features. SEs typically regulate key genes that can determine cell identity and differentiation. Identifying SEs and their effects may be critical in predicting key regulatory genes, such as master transcription factor genes or oncogenes. Signal inducible SEs are dense stretches of signal terminal transcription factor (TF) binding regions, and may modulate the interaction between environmental factors (e.g., Vitamin D) and genetic factors (i.e., risk variants) in complex diseases such as multiple sclerosis (MS). As a complex autoimmune disease, the etiology and progression of MS, including the interaction between Vitamin D and MS risk variants, is still unclear and can be explored from the aspect of signal SEs. Vitamin D [with its active form: 1,25(OH)2D3], is an environmental risk factor for MS. It binds the Vitamin D receptor (VDR) and regulates gene expression. This study explores the association between VDR super-enhancers (VSEs) and MS risk variants. Firstly, we reanalyse public ChIP-seq and RNA-seq data to classify VSEs into three categories according to their combinations of persistent and secondary VDR binding. Secondly, we indicate the genes with VSE regions that are near MS risk variants. Furthermore, we find that MS risk variants are enriched in VSE regions, and we indicate some genes with a VSE overlapping MS risk variant for further exploration. We also find two clusters of genes from the set of genes showing correlation of expression patterns with the MS risk gene ZMIZ1 that appear to be regulated by VSEs in THP-1 cells. It is the first time that VSEs have been analyzed, and we directly connect the genetic risk factors for MS risk with Vitamin D based on VSEs.
Collapse
Affiliation(s)
- Ming Lu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.,Department of Immunology, Anhui Medical University, Hefei, China
| | - Bennet J McComish
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Bruce V Taylor
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Heinrich Körner
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.,Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
37
|
Parameters of Bone and Cardiovascular Health Related to 25-Hydroxyvitamin D Status in Emirati Nationals attending Primary Care and Diabetes services: a retrospective cohort study. Sci Rep 2019; 9:3835. [PMID: 30846793 PMCID: PMC6405844 DOI: 10.1038/s41598-019-40523-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 02/14/2019] [Indexed: 12/19/2022] Open
Abstract
Vitamin D deficiency is endemic in people living in the Gulf states. We performed a retrospective analysis of data gathered at the first attendance of 82,396 Emirati nationals to outpatient diabetes, endocrinology and general primary care services at two centres in the United Arab Emirates during 2012–2016. Our aim was to explore associations between vitamin D status and markers of cardiovascular and bone health. In the study population, 67.1% of men and 73.5% of women had serum 25(OH)D of less than 50 nmol/L, with the lowest levels being found in young adults. Among Emirati adults with type 2 diabetes, serum 25(OH)D < 50 nmol/L was associated with an increased risk of a coexisting adverse total cholesterol:HDL (TC:HDL) ratio (odds ratio 2.13 (1.60–2.84), p < 0.001). Correcting for age, sex, body mass index, HbA1c and statin therapy, an increase in 25(OH)D of 1 nmol/L was associated with a 0.01 unit reduction in TC:HDL in this population. In a subset of 1064 adult individuals, 25(OH)D < 25 nmol/L was associated with a reduction in DEXA-measured z-score of −0.29 (−0.44 to −0.15, p < 0.001) at the femoral neck and of −0.25(−0.45 to −0.05, p = 0.015) at L1–4, corrected for body mass index, compared with individuals with 25(OH)D ≥ 75 nmol/L. Our findings raise concerns regarding lifetime burden of cardiovascular disease and bone health for young Emiratis with vitamin D deficiency.
Collapse
|
38
|
Nurminen V, Seuter S, Carlberg C. Primary Vitamin D Target Genes of Human Monocytes. Front Physiol 2019; 10:194. [PMID: 30890957 PMCID: PMC6411690 DOI: 10.3389/fphys.2019.00194] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/15/2019] [Indexed: 12/31/2022] Open
Abstract
The molecular basis of vitamin D signaling implies that the metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) of the secosteroid vitamin D3 activates the transcription factor vitamin D receptor (VDR), which in turn modulates the expression of hundreds of primary vitamin D target genes. Since the evolutionary role of nuclear receptors, such as VDR, was the regulation of cellular metabolism, the control of calcium metabolism became the primary function of vitamin D and its receptor. Moreover, the nearly ubiquitous expression of VDR enabled vitamin D to acquire additional physiological functions, such as the support of the innate immune system in its defense against microbes. Monocytes and their differentiated phenotypes, macrophages and dendritic cells, are key cell types of the innate immune system. Vitamin D signaling was most comprehensively investigated in THP-1 cells, which are an established model of human monocytes. This includes the 1,25(OH)2D3-modulated cistromes of VDR, the pioneer transcription factors PU.1 and CEBPA and the chromatin modifier CTCF as well as of the histone markers of promoter and enhancer regions, H3K4me3 and H3K27ac, respectively. These epigenome-wide datasets led to the development of our chromatin model of vitamin D signaling. This review discusses the mechanistic basis of 189 primary vitamin D target genes identified by transcriptome-wide analysis of 1,25(OH)2D3-stimulated THP-1 cells and relates the epigenomic basis of four different regulatory scenarios to the physiological functions of the respective genes.
Collapse
Affiliation(s)
- Veijo Nurminen
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Sabine Seuter
- Institute for Cardiovascular Physiology, Medical Faculty, Goethe University Frankfurt, Frankfurt, Germany
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
39
|
Tuckey RC, Cheng CYS, Slominski AT. The serum vitamin D metabolome: What we know and what is still to discover. J Steroid Biochem Mol Biol 2019; 186:4-21. [PMID: 30205156 PMCID: PMC6342654 DOI: 10.1016/j.jsbmb.2018.09.003] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/04/2018] [Accepted: 09/04/2018] [Indexed: 01/08/2023]
Abstract
Vitamin D, referring to the two forms, D2 from the diet and D3 primarily derived from phototransformation in the skin, is a prohormone important in human health. The most hormonally active form, 1α,25-dihydroxyvitamin D (1α,25(OH)2D), formed from vitamin D via 25-hydroxyvitamin D (25(OH)D), is not only important for regulating calcium metabolism, but has many pleiotropic effects including regulation of the immune system and has anti-cancer properties. The major circulating form of vitamin D is 25(OH)D and both D2 and D3 forms are routinely measured by LC/MS/MS to assess vitamin D status, due to their relatively long half-lives and much higher concentrations compared to 1α,25(OH)2D. Inactivation of both 25(OH)D and 1α,25(OH)2D is catalyzed by CYP24A1 and 25-hydroxyvitamin D3 3-epimerase. Initial products from these enzymes acting on 25(OH)D3 are 24R,25(OH)2D3 and 3-epi-25(OH)D3, respectively, and both of these can also be measured routinely in some clinical laboratories to further document vitamin D status. With advances in LC/MS/MS and its increased availability, and with the help of studies with recombinant vitamin D-metabolizing enzymes, many other vitamin D metabolites have now been detected and in some cases quantitated, in human serum. CYP11A1 which catalyzes the first step in steroidogenesis, has been found to also act on vitamins D3 and D2 hydroxylating both at C20, but with some secondary metabolites produced by subsequent hydroxylations at other positions on the side chain. The major vitamin D3 metabolite, 20S-hydroxyvitamin D3 (20S(OH)D3), shows biological activity, often similar to 1α,25(OH)2D3 but without calcemic effects. Using standards produced enzymatically by purified CYP11A1 and characterized by NMR, many of these new metabolites have been detected in human serum, with semi-quantitative measurement of 20S(OH)D3 indicating it is present at comparable concentrations to 24R,25(OH)2D3 and 3-epi-25(OH)D3. Recently, vitamin D-related hydroxylumisterols derived from lumisterol3, a previtamin D3 photoproduct, have also been measured in human serum and displayed biological activity in initial in vitro studies. With the current extensive knowledge on the reactions and pathways of metabolism of vitamin D, especially those catalyzed by CYP24A1, CYP27A1, CYP27B1, CYP3A4 and CYP11A1, it is likely that many other of the resulting hydroxyvitamin D metabolites will be measured in human serum in the future, some contributing to a more detailed understanding of vitamin D status in health and disease.
Collapse
Affiliation(s)
- Robert C Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA, 6009, Australia.
| | - Chloe Y S Cheng
- School of Molecular Sciences, The University of Western Australia, Perth, WA, 6009, Australia
| | - Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, AL, 35294, USA; Comprehensive Cancer Center Cancer Chemoprevention Program, University of Alabama at Birmingham, AL, 35294, USA; VA Medical Center, Birmingham, AL, 35294, USA
| |
Collapse
|
40
|
Carlberg C, Neme A. Machine learning approaches infer vitamin D signaling: Critical impact of vitamin D receptor binding within topologically associated domains. J Steroid Biochem Mol Biol 2019; 185:103-109. [PMID: 30044963 DOI: 10.1016/j.jsbmb.2018.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/17/2018] [Accepted: 07/21/2018] [Indexed: 12/28/2022]
Abstract
The vitamin D-modulated transcriptome of highly responsive human cells, such as THP-1 monocytes, comprises more than 500 genes, half of which are primary targets. Recently, we proposed a chromatin model of vitamin D signaling demonstrating that nearly all vitamin D target genes are located within vitamin D-modulated topologically associated domains (TADs). This model is based on genome-wide binding patterns of the vitamin D receptor (VDR), the pioneer transcription factor PU.1, the chromatin organizer CTCF and histone markers of active promoter regions (H3K4me3) and active chromatin (H3K27ac). In addition, time-dependent data on accessible chromatin and mRNA expression are implemented. For the interrogation and in deep inspection of these high-dimensional datasets unsupervised and supervised machine learning algorithms were applied. Unsupervised methods, such as the vector quantization tool K-means and the dimensionality reduction algorithm self-organizing map, generated descriptions of how attributes, such as VDR binding and chromatin accessibility, affect each other as a function of time and/or co-localization within the same genomic region. Supervised algorithms, such as random forests, allowed the data to be classified into pre-existing categories like persistent (i.e. constant) and time-dependent (i.e. transient) VDR binding sites. The relative amounts of these VDR categories in TADs showed to be the main discriminator for sorting the latter into five classes carrying vitamin D target genes involved in distinct biological processes. In conclusion, via the application of machine learning methods we identified the spatio-temporal VDR binding pattern in TADs as the most critical attribute for specific regulation of vitamin D target genes and the segregation of vitamin D's physiologic function.
Collapse
Affiliation(s)
- Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FIN-70211 Kuopio, Finland.
| | - Antonio Neme
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FIN-70211 Kuopio, Finland
| |
Collapse
|
41
|
Nurminen V, Neme A, Seuter S, Carlberg C. Modulation of vitamin D signaling by the pioneer factor CEBPA. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:96-106. [PMID: 30550771 DOI: 10.1016/j.bbagrm.2018.12.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 01/31/2023]
Abstract
The myeloid master regulator CCAAT enhancer-binding protein alpha (CEBPA) is known as a pioneer factor. In this study, we report the CEBPA cistrome of THP-1 human monocytes after stimulation with the vitamin D receptor (VDR) ligand 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) for 2, 8 and 24 h. About a third of the genomic VDR binding sites co-located with those of CEBPA. In parallel, the binding strength of 5% of the CEBPA cistrome, i.e. some 1500 sites, is significantly (p < 0.001) affected by 1,25(OH)2D3. Transcriptome-wide analysis after CEBPA silencing indicated that the pioneer factor enhances both the basal expression and ligand inducibility of 70 vitamin D target genes largely involved in lipid signaling and metabolism. In contrast, CEBPA suppresses 82 vitamin D target genes many of which are related to the modulation of T cell activity by monocytes. The inducibility of the promoter-specific histone marker H3K4me3 distinguishes the former class of genes from the latter. Moreover, prominent occupancy of the myeloid pioneer factor PU.1 on 1,25(OH)2D3-sensitive CEBPA enhancers mechanistically explains the dichotomy of vitamin D target genes. In conclusion, CEBPA supports vitamin D signaling concerning actions of the innate immune system, but uses the antagonism with PU.1 for suppressing possible overreactions of adaptive immunity.
Collapse
Affiliation(s)
- Veijo Nurminen
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Antonio Neme
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Sabine Seuter
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland.
| |
Collapse
|
42
|
Slominski AT, Kim TK, Janjetovic Z, Brożyna AA, Żmijewski MA, Xu H, Sutter TR, Tuckey RC, Jetten AM, Crossman DK. Differential and Overlapping Effects of 20,23(OH)₂D3 and 1,25(OH)₂D3 on Gene Expression in Human Epidermal Keratinocytes: Identification of AhR as an Alternative Receptor for 20,23(OH)₂D3. Int J Mol Sci 2018; 19:ijms19103072. [PMID: 30297679 PMCID: PMC6213311 DOI: 10.3390/ijms19103072] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/28/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022] Open
Abstract
A novel pathway of vitamin D activation by CYP11A has previously been elucidated. To define the mechanism of action of its major dihydroxy-products, we tested the divergence and overlap between the gene expression profiles of human epidermal keratinocytes treated with either CYP11A1-derived 20,23(OH)2D3 or classical 1,25(OH)2D3. Both secosteroids have significant chemical similarity with the only differences being the positions of the hydroxyl groups. mRNA was isolated and examined by microarray analysis using Illumina’s HumanWG-6 chip/arrays and subsequent bioinformatics analyses. Marked differences in the up- and downregulated genes were observed between 1,25(OH)2D3- and 20,23(OH)2D3-treated cells. Hierarchical clustering identified both distinct, opposite and common (overlapping) gene expression patterns. CYP24A1 was a common gene strongly activated by both compounds, a finding confirmed by qPCR. Ingenuity pathway analysis identified VDR/RXR signaling as the top canonical pathway induced by 1,25(OH)2D3. In contrast, the top canonical pathway induced by 20,23(OH)2D3 was AhR, with VDR/RXR being the second nuclear receptor signaling pathway identified. QPCR analyses validated the former finding by revealing that 20,23(OH)2D3 stimulated CYP1A1 and CYP1B1 gene expression, effects located downstream of AhR. Similar stimulation was observed with 20(OH)D3, the precursor to 20,23(OH)2D3, as well as with its downstream metabolite, 17,20,23(OH)3D3. Using a Human AhR Reporter Assay System we showed marked activation of AhR activity by 20,23(OH)2D3, with weaker stimulation by 20(OH)D3. Finally, molecular modeling using an AhR LBD model predicted vitamin D3 hydroxyderivatives to be good ligands for this receptor. Thus, our microarray, qPCR, functional studies and molecular modeling indicate that AhR is the major receptor target for 20,23(OH)2D3, opening an exciting area of investigation on the interaction of different vitamin D3-hydroxyderivatives with AhR and the subsequent downstream activation of signal transduction pathways in a cell-type-dependent manner.
Collapse
Affiliation(s)
- Andrzej T Slominski
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Comprehensive Cancer Center, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Veteran Administration Medical Center, Birmingham, AL 35294, USA.
| | - Tae-Kang Kim
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Zorica Janjetovic
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Anna A Brożyna
- Department of Medical Biology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 87-100 Toruń, Poland.
- Department of Tumor Pathology and Pathomorphology, Oncology Centre-Prof. Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland.
| | - Michal A Żmijewski
- Department of Histology, Medical University of Gdańsk, 80-211 Gdańsk, Poland.
| | - Hui Xu
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Thomas R Sutter
- Feinstone Center for Genomic Research, University of Memphis, Memphis, TN 38152 USA.
| | - Robert C Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia.
| | - Anton M Jetten
- Immunity, Inflammation, and Disease Laboratory/Cell Biology Group, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA.
| | - David K Crossman
- Howell and Elizabeth Heflin Center for Human Genetics, Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
43
|
Nurminen V, Neme A, Seuter S, Carlberg C. The impact of the vitamin D-modulated epigenome on VDR target gene regulation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:697-705. [PMID: 30018005 DOI: 10.1016/j.bbagrm.2018.05.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 01/12/2023]
Abstract
The micronutrient vitamin D significantly modulates the human epigenome via enhancing genome-wide the rate of accessible chromatin and vitamin D receptor (VDR) binding. This study focuses on histone marks of active chromatin at promoter and enhancer regions and investigates, whether these genomic loci are sensitive to vitamin D. The epigenome of THP-1 human monocytes contains nearly 23,000 sites with H3K4me3 histone modifications, 550 of which sites are significantly (p < 0.05) modulated by stimulation with the VDR ligand 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3). H3K27ac histone modifications mark active chromatin and 2473 of 45,500 sites are vitamin D sensitive. The two types of ligand-dependent histone marks allow to distinguish promoter and enhancer regulation by vitamin D, respectively. Transcription start site overlap is the prime attribute of ligand-dependent H3K4me3 marks, while VDR co-location is the top ranking parameter describing 1,25(OH)2D3-sensitive H3K27ac marks at enhancers. A categorization of 1,25(OH)2D3-sensitive histone marks by machine learning algorithms - using the attributes overall peak strength and ligand inducibility - highlights 260 and 287 regions with H3K4me3 and H3K27ac modifications, respectively. These loci are found at the promoter regions of 59 vitamin D target genes and their associated enhancers. In this way, ligand-dependent histone marks provide a link of the effects of 1,25(OH)2D3 on the epigenome with previously reported mRNA expression changes of vitamin D target genes. In conclusion, the human epigenome responds also on the level of histone modifications to 1,25(OH)2D3 stimulation. This allows a more detailed understanding of vitamin D target gene regulation.
Collapse
Affiliation(s)
- Veijo Nurminen
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Antonio Neme
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Sabine Seuter
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211 Kuopio, Finland.
| |
Collapse
|
44
|
Lu M, Taylor BV, Körner H. Genomic Effects of the Vitamin D Receptor: Potentially the Link between Vitamin D, Immune Cells, and Multiple Sclerosis. Front Immunol 2018; 9:477. [PMID: 29593729 PMCID: PMC5857605 DOI: 10.3389/fimmu.2018.00477] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/22/2018] [Indexed: 12/12/2022] Open
Abstract
Vitamin D has a plethora of functions that are important for the maintenance of general health and in particular, the functional integrity of the immune system, such as promoting an anti-inflammatory cytokine profile and reducing the Treg/Th17 ratio. Multiple sclerosis (MS) is a chronic, inflammatory, and neurodegenerative central nervous system (CNS) disorder of probable autoimmune origin. MS is characterized by recurring or progressive demyelination and degeneration of the CNS due in part to a misguided immune response to as yet undefined (CNS) antigens, potentially including myelin basic protein and proteolipid protein. MS has also been shown to be associated significantly with environmental factors such as the lack of vitamin D. The role of vitamin D in the pathogenesis and progression of MS is complex. Recent genetic studies have shown that various common MS-associated risk-single-nucleotide polymorphisms (SNPs) are located within or in the vicinity of genes associated with the complex metabolism of vitamin D. The functional aspects of these genetic associations may be explained either by a direct SNP-associated loss- or gain-of-function in a vitamin D-associated gene or due to a change in the regulation of gene expression in certain immune cell types. The development of new genetic tools using next-generation sequencing: e.g., chromatin immunoprecipitation sequencing (ChIP-seq) and the accompanying rapid progress of epigenomics has made it possible to recognize that the association between vitamin D and MS could be based on the extensive and characteristic genomic binding of the vitamin D receptor (VDR). Therefore, it is important to analyze comprehensively the spatiotemporal VDR binding patterns that have been identified using ChIP-seq in multiple immune cell types to reveal an integral profile of genomic VDR interaction. In summary, the aim of this review is to connect genomic effects vitamin D has on immune cells with MS and thus, to contribute to a better understanding of the influence of vitamin D on the etiology and the pathogenesis of this complex autoimmune disease.
Collapse
Affiliation(s)
- Ming Lu
- Menzies Institute for Medical Research Tasmania, Hobart, TAS, Australia
| | - Bruce V. Taylor
- Menzies Institute for Medical Research Tasmania, Hobart, TAS, Australia
| | - Heinrich Körner
- Menzies Institute for Medical Research Tasmania, Hobart, TAS, Australia
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Engineering Technology Research Center of Anti-inflammatory and Immunodrugs in Anhui Province, Hefei, China
| |
Collapse
|
45
|
Seuter S, Neme A, Carlberg C. ETS transcription factor family member GABPA contributes to vitamin D receptor target gene regulation. J Steroid Biochem Mol Biol 2018; 177:46-52. [PMID: 28870774 DOI: 10.1016/j.jsbmb.2017.08.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/05/2017] [Accepted: 08/08/2017] [Indexed: 01/31/2023]
Abstract
Binding motifs of the ETS-domain transcription factor GABPA are found with high significance below the summits of the vitamin D receptor (VDR) cistrome. VDR is the nuclear receptor for the biologically most active vitamin D metabolite 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3). In this study, we determined the GABPA cistrome in THP-1 human monocytes and found that it is comprised of 3822 genomic loci, some 20% of which were modulated by 1,25(OH)2D3. The GABPA cistrome showed a high overlap rate with accessible chromatin and the pioneer transcription factor PU.1. Interestingly, 23 and 12% of persistent and transient VDR binding sites, respectively, co-localized with GABPA, which is clearly higher than the rate of secondary VDR loci (4%). Some 40% of GABPA binding sites were found at transcription start sites, nearly 100 of which are of 1,25(OH)2D3 target genes. On 593 genomic loci VDR and GABPA co-localized with PU.1, while only 175 VDR sites bound GABPA in the absence of PU.1. In total, VDR sites with GABPA co-localization may control some 450 vitamin D target genes. Those genes that are co-controlled by PU.1 preferentially participate in cellular and immune signaling processes, while the remaining genes are involved in cellular metabolism pathways. In conclusion, GABPA may contribute to differential VDR target gene regulation.
Collapse
Affiliation(s)
- Sabine Seuter
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211, Kuopio, Finland
| | - Antonio Neme
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211, Kuopio, Finland
| | - Carsten Carlberg
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, FI-70211, Kuopio, Finland.
| |
Collapse
|
46
|
Abstract
The vitamin D3 metabolite 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3] is the exclusive high-affinity ligand of the vitamin D receptor (VDR), a transcription factor with direct effects on gene expression. Transcriptome- and epigenome-wide data obtained in THP-1 human monocytes are the basis of the chromatin model of vitamin D signaling. The model describes, how VDR's spatio-temporal binding profile provides key insight into the pleiotropic action of vitamin D. The transcription of some 300 primary target genes is significantly modulated through the action of genomic VDR binding sites in concert with the pioneer transcription factor PU.1 and the chromatin organizer CTCF. In parallel, the short-term vitamin D intervention study VitDbol (NCT02063334) was designed, in order to extrapolate insight into vitamin D signaling from in vitro to in vivo. Before and 24 h after a vitamin D3 bolus chromatin and RNA were prepared from peripheral blood mononuclear cells for epigenome- and transcriptome-wide analysis. The study subjects showed a personalized response to vitamin D and could be distinguished into high, mid, and low responders. Comparable principles of vitamin D signaling were identified in vivo and in vitro concerning target gene responses as well as changes in chromatin accessibility. In conclusion, short-term vitamin D supplementation studies represent a new type of safe in vivo investigations demonstrating that vitamin D and its metabolites have direct effects on the human epigenome and modulate the response of the transcriptome in a personalized fashion.
Collapse
|