Vitamin D receptor cistrome-transcriptome analyses establishes quantitatively distinct receptor genomic interactions in African American prostate cancer regulated by BAZ1A

Background African American (AA) prostate cancer (PCa) appears uniquely sensitive to 1α,25(OH)2D3 signaling, compared to European American (EA) PCa, but the extent and impact of vitamin D receptor genomic functions remain poorly defined. Results A panel of EA and AA prostate epithelial cells (EA: HPr1-AR, LNCaP, AA: RC43N, RC43T, RC77N, RC77T) were analyzed with RIME to reveal the cell-specific composition of the VDR- complex. 1α,25(OH)2D3-dependent ATAC-Seq revealed the greatest impact on nucleosome positioning in RC43N and RC43T, with gain of nucleosome-free at enhancer regions. VDR ChIP-Seq identified stronger and more frequent VDR binding in RC43N and RC43T that was enriched for a larger and distinct motif repertoire, than EA cells. VDR binding significantly overlapped with core circadian rhythm transcription factors in AA cell line models. RNA-Seq also revealed significantly stronger 1α,25(OH)2D3 dependent VDR transcriptional responses enriched for circadian rhythm and inflammation networks in AA cells. Whilst RC43N was most responsive, RC43T displayed distorted responses. Significantly reduced BAZ1A/SMARCA5 in AA PCa samples was identified, and restored BAZ1A expression uniquely and significantly increased 1α,25(OH)2D3-regulated VDR targets in AA cells. These VDR- dependent cistrome-annotated genes were also uniquely and most significantly identified in three cohorts of AA PCa patients. Conclusion These data suggest VDR transcriptional control in the prostate is more potent and dynamic in AA men, and primed to govern inflammatory and circadian pathways. Reduced BAZ1A/SMARCA5 expression and/or reduced environmentally-regulated serum vitamin D3 levels suppress these actions. Therefore, the VDR axis lies at the cross-roads of biopsychosocial processes including stress responses, access to quality early detection and treatment, social determinants and that collectively contribute to PCa health disparities.

[1]  J. Manson,et al.  Vitamin D and marine omega 3 fatty acid supplementation and incident autoimmune disease: VITAL randomized controlled trial , 2022, British medical journal.

[2]  Sahar J Farahani,et al.  Prostate tumors of native men from West Africa show biologically distinct pathways—A comparative genomic study , 2021, The Prostate.

[3]  Minlu Zhang,et al.  Phase Ib study of patients with metastatic castrate-resistant prostate cancer treated with different sequencing regimens of atezolizumab and sipuleucel-T , 2021, Journal for ImmunoTherapy of Cancer.

[4]  M. Quek,et al.  African American Men have Increased Risk of Prostate Cancer Detection despite Similar Rates of Anterior Prostatic Lesions and PI-RADS Grade on Multiparametric Magnetic Resonance Imaging. , 2021, Urology.

[5]  Randy V. Bradley,et al.  Comparative analysis of 1152 African-American and European-American men with prostate cancer identifies distinct genomic and immunological differences , 2021, Communications biology.

[6]  J. Manson,et al.  The VITamin D and OmegA-3 TriaL (VITAL): Do Results Differ by Sex or Race/Ethnicity? , 2020, American journal of lifestyle medicine.

[7]  E. Mathé,et al.  Identification of transcription factor co-regulators that drive prostate cancer progression , 2020, Scientific Reports.

[8]  J. Manson,et al.  Effect of Vitamin D3 Supplements on Development of Advanced Cancer , 2020, JAMA network open.

[9]  Jianfeng Xu,et al.  Distinct Genomic Alterations in Prostate Tumors Derived from African American Men , 2020, Molecular Cancer Research.

[10]  Joshua D. Campbell,et al.  Comparative Genomics Reveals Distinct Immune-oncologic Pathways in African American Men with Prostate Cancer , 2020, Clinical Cancer Research.

[11]  Joshua D. Campbell,et al.  Genomic Profiling of Prostate Cancers from Men with African and European Ancestry , 2020, Clinical Cancer Research.

[12]  H. Wheeler,et al.  Multi-ethnic transcriptome-wide association study of prostate cancer , 2020, bioRxiv.

[13]  Chang-Hoon Kim,et al.  Relationship between Sleep Duration, Sun Exposure, and Serum 25-Hydroxyvitamin D Status: A Cross-sectional Study , 2020, Scientific Reports.

[14]  Clifford A. Meyer,et al.  Lisa: inferring transcriptional regulators through integrative modeling of public chromatin accessibility and ChIP-seq data , 2020, Genome Biology.

[15]  T. Rebbeck,et al.  Integrative comparison of the genomic and transcriptomic landscape between prostate cancer patients of predominantly African or European genetic ancestry , 2020, PLoS genetics.

[16]  Bing Ren,et al.  A Compendium of Promoter-Centered Long-Range Chromatin Interactions in the Human Genome , 2019, Nature Genetics.

[17]  L. Arab,et al.  The Prostate , 2019 .

[18]  Hannah Stower Cancer risk , 2019, Nature Medicine.

[19]  C. Carlberg,et al.  Vitamin D Signaling Suppresses Early Prostate Carcinogenesis in TgAPT121 Mice , 2019, Cancer Prevention Research.

[20]  L. Nonn,et al.  High levels of PIWI‐interacting RNAs are present in the small RNA landscape of prostate epithelium from vitamin D clinical trial specimens , 2019, The Prostate.

[21]  S. Shimba,et al.  Intestinal clock system regulates skeletal homeostasis. , 2019, JCI insight.

[22]  R. Kittles,et al.  Genetic Ancestry Analysis Reveals Misclassification of Commonly Used Cancer Cell Lines , 2019, Cancer Epidemiology, Biomarkers & Prevention.

[23]  R. Kittles,et al.  Physiologic serum 1,25 dihydroxyvitamin D is inversely associated with prostatic Ki67 staining in a diverse sample of radical prostatectomy patients , 2019, Cancer Causes & Control.

[24]  J. Manson,et al.  Marine n‐3 Fatty Acids and Prevention of Cardiovascular Disease and Cancer , 2019, The New England journal of medicine.

[25]  Lara E Sucheston-Campbell,et al.  The miR-96 and RARγ signaling axis governs androgen signaling and prostate cancer progression , 2018, Oncogene.

[26]  Gordon K Smyth,et al.  The R package Rsubread is easier, faster, cheaper and better for alignment and quantification of RNA sequencing reads , 2018, bioRxiv.

[27]  Andrew N. Holding,et al.  A quantitative mass spectrometry-based approach to monitor the dynamics of endogenous chromatin-associated protein complexes , 2018, Nature Communications.

[28]  D. Trump Calcitriol and cancer therapy: A missed opportunity , 2018, Bone reports.

[29]  Jun Yu,et al.  ATACseqQC: a Bioconductor package for post-alignment quality assessment of ATAC-seq data , 2018, BMC Genomics.

[30]  Kathryn S. Burch,et al.  Leveraging polygenic functional enrichment to improve GWAS power , 2017, bioRxiv.

[31]  R. Matsunuma,et al.  Phosphorylated HBO1 at UV irradiated sites is essential for nucleotide excision repair , 2017, Nature Communications.

[32]  S. Mayne,et al.  Serum 25‐hydroxyvitamin D, vitamin D binding protein, and prostate cancer risk in black men , 2017, Cancer.

[33]  Brent S. Pedersen,et al.  GIGGLE: a search engine for large-scale integrated genome analysis , 2017, Nature Methods.

[34]  K. Lunetta,et al.  Demographic, lifestyle, and genetic determinants of circulating concentrations of 25-hydroxyvitamin D and vitamin D-binding protein in African American and European American women. , 2017, The American journal of clinical nutrition.

[35]  R. Kittles,et al.  Prostatic compensation of the vitamin D axis in African American men. , 2017, JCI insight.

[36]  E. Jacobs,et al.  Race and BMI modify associations of calcium and vitamin D intake with prostate cancer , 2017, BMC Cancer.

[37]  R. Kittles,et al.  Association between Serum 25-Hydroxy-Vitamin D and Aggressive Prostate Cancer in African American Men , 2016, Nutrients.

[38]  K. Dear,et al.  Investigating the patterns and determinants of seasonal variation in vitamin D status in Australian adults: the Seasonal D Cohort Study , 2016, BMC Public Health.

[39]  R. Kittles,et al.  Can vitamin D supplementation reduce prostate cancer disparities? , 2016, Pharmacogenomics.

[40]  G. Hardiman,et al.  Systems analysis of the prostate transcriptome in African-American men compared with European-American men. , 2016, Pharmacogenomics.

[41]  T. Visakorpi,et al.  Inhibition of the glucocorticoid receptor results in an enhanced miR-99a/100-mediated radiation response in stem-like cells from human prostate cancers , 2016, Oncotarget.

[42]  Lara E Sucheston-Campbell,et al.  Genetic variations in vitamin D‐related pathways and breast cancer risk in African American women in the AMBER consortium , 2016, International journal of cancer.

[43]  Aaron T. L. Lun,et al.  Three-dimensional disorganization of the cancer genome occurs coincident with long-range genetic and epigenetic alterations , 2016, Genome research.

[44]  R. Kittles,et al.  Vitamin D and Immune Response: Implications for Prostate Cancer in African Americans , 2016, Front. Immunol..

[45]  J. Carroll,et al.  Rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) for analysis of chromatin complexes , 2016, Nature Protocols.

[46]  Aaron T. L. Lun,et al.  csaw: a Bioconductor package for differential binding analysis of ChIP-seq data using sliding windows , 2015, Nucleic acids research.

[47]  Ellen T. Gelfand,et al.  A Novel Approach to High-Quality Postmortem Tissue Procurement: The GTEx Project , 2015, Biopreservation and biobanking.

[48]  Pui-Yan Kwok,et al.  A large multiethnic genome-wide association study of prostate cancer identifies novel risk variants and substantial ethnic differences. , 2015, Cancer discovery.

[49]  T. Naab,et al.  Risk of prostate cancer in African‐American men: Evidence of mixed effects of dietary quercetin by serum vitamin D status , 2015, The Prostate.

[50]  James L. Mohler,et al.  Associations of prostate cancer risk variants with disease aggressiveness: results of the NCI-SPORE Genetics Working Group analysis of 18,343 cases , 2015, Human Genetics.

[51]  Michael Q. Zhang,et al.  Integrative analysis of 111 reference human epigenomes , 2015, Nature.

[52]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[53]  J. Brodbelt,et al.  Screen identifies bromodomain protein ZMYND8 in chromatin recognition of transcription-associated DNA damage that promotes homologous recombination , 2015, Genes & development.

[54]  D. DeFranco,et al.  VDR Activity Is Differentially Affected by Hic-5 in Prostate Cancer and Stromal Cells , 2014, Molecular Cancer Research.

[55]  R. Kittles,et al.  Vitamin D Deficiency Predicts Prostate Biopsy Outcomes , 2014, Clinical Cancer Research.

[56]  Hanfei Sun,et al.  Target analysis by integration of transcriptome and ChIP-seq data with BETA , 2013, Nature Protocols.

[57]  Yan Li,et al.  A high-resolution map of three-dimensional chromatin interactome in human cells , 2013, Nature.

[58]  N. Keating,et al.  Ultraviolet index and racial differences in prostate cancer incidence and mortality , 2013, Cancer.

[59]  J. Vadgama,et al.  Vitamin D Receptor Gene Polymorphisms and Prognosis of Breast Cancer among African-American and Hispanic Women , 2013, PloS one.

[60]  D. Smiraglia,et al.  Recruitment of NCOR1 to VDR target genes is enhanced in prostate cancer cells and associates with altered DNA methylation patterns. , 2013, Carcinogenesis.

[61]  M. Campbell,et al.  Vitamin D receptor signaling mechanisms: Integrated actions of a well-defined transcription factor , 2013, Steroids.

[62]  J. Schalken,et al.  DC‐SCRIPT: AR and VDR regulator lost upon transformation of prostate epithelial cells , 2012, The Prostate.

[63]  E. Garrett-Mayer,et al.  Vitamin D3 supplementation at 4000 international units per day for one year results in a decrease of positive cores at repeat biopsy in subjects with low-risk prostate cancer under active surveillance. , 2012, The Journal of clinical endocrinology and metabolism.

[64]  R. Kittles,et al.  Predictors of Serum Vitamin D Levels in African American and European American Men in Chicago , 2012, American journal of men's health.

[65]  N. Jablonski The evolution of human skin colouration and its relevance to health in the modern world. , 2012, The journal of the Royal College of Physicians of Edinburgh.

[66]  D. Alberts,et al.  Phase III Trial of Selenium to Prevent Prostate Cancer in Men with High-grade Prostatic Intraepithelial Neoplasia: SWOG S9917 , 2011, Cancer Prevention Research.

[67]  M. Rubin,et al.  TMPRSS2–ERG gene fusion prevalence and class are significantly different in prostate cancer of caucasian, african‐american and japanese patients , 2011, The Prostate.

[68]  Assam El-Osta,et al.  Clonogenic assay: adherent cells. , 2011, Journal of visualized experiments : JoVE.

[69]  J. Rhim,et al.  Establishment and characterization of a pair of non-malignant and malignant tumor derived cell lines from an African American prostate cancer patient. , 2010, International journal of oncology.

[70]  B. Turner,et al.  Epigenetic control of a VDR-governed feed-forward loop that regulates p21(waf1/cip1) expression and function in non-malignant prostate cells , 2010, Nucleic acids research.

[71]  A. Templeton,et al.  Evolutionary perspective in skin color, vitamin D and its receptor , 2010, Hormones.

[72]  C. Cooper,et al.  Elevated NCOR1 disrupts PPARalpha/gamma signaling in prostate cancer and forms a targetable epigenetic lesion. , 2010, Carcinogenesis.

[73]  Zhentao Zhang,et al.  Constitutive activation of the mitogen‐activated protein kinase pathway impairs vitamin D signaling in human prostate epithelial cells , 2010, Journal of cellular physiology.

[74]  S. Heikkinen,et al.  The down-regulation of the human MYC gene by the nuclear hormone 1alpha,25-dihydroxyvitamin D3 is associated with cycling of corepressors and histone deacetylases. , 2010, Journal of molecular biology.

[75]  B. Diffey Modelling the seasonal variation of vitamin D due to sun exposure , 2010, The British journal of dermatology.

[76]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[77]  R. Baumgartner,et al.  Serum Vitamin D and Breast Density in Breast Cancer Survivors , 2010, Cancer Epidemiology, Biomarkers & Prevention.

[78]  F. Campbell,et al.  The Yin and Yang of vitamin D receptor (VDR) signaling in neoplastic progression: Operational networks and tissue-specific growth control , 2010, Biochemical pharmacology.

[79]  F. Delaunay,et al.  The nuclear hormone receptor family round the clock. , 2008, Molecular endocrinology.

[80]  M. Gleave,et al.  Association of TMPRSS2-ERG gene fusion with clinical characteristics and outcomes: results from a population-based study of prostate cancer , 2008, BMC Cancer.

[81]  S. Ingles,et al.  Sun exposure, vitamin D receptor gene polymorphisms, and breast cancer risk in a multiethnic population. , 2007, American journal of epidemiology.

[82]  D. Trump,et al.  Phase II trial of high‐dose, intermittent calcitriol (1,25 dihydroxyvitamin D3) and dexamethasone in androgen‐independent prostate cancer , 2006, Cancer.

[83]  J. Tchinda,et al.  Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. , 2006, Science.

[84]  J. Tchinda,et al.  Recurrent Fusion of TMPRSS2 and ETS Transcription Factor Genes in Prostate Cancer , 2005, Science.

[85]  R. Kittles,et al.  Sequence variation within the 5′ regulatory regions of the vitamin D binding protein and receptor genes and prostate cancer risk , 2005, The Prostate.

[86]  F. Khanim,et al.  Altered SMRT levels disrupt vitamin D3 receptor signalling in prostate cancer cells , 2004, Oncogene.

[87]  M. Tyers,et al.  The GRID: The General Repository for Interaction Datasets , 2003, Genome Biology.

[88]  M. Ling,et al.  Androgen induces differentiation of a human papillomavirus 16 E6/E7 immortalized prostate epithelial cell line. , 2001, The Journal of endocrinology.

[89]  D. Feldman,et al.  1,25-dihydroxyvitamin D3 and malignant melanoma: the presence of receptors and inhibition of cell growth in culture. , 1981, Endocrinology.

[90]  A. Jemal,et al.  Incidence and Mortality , 2011 .

[91]  C. Coopersmith,et al.  Epithelial cells , 1991 .